Language selection

Search

Patent 2548180 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2548180
(54) English Title: METHODS OF KILLING TUMOR CELLS BY TARGETING INTERNAL ANTIGENS EXPOSED ON APOPTOTIC TUMOR CELLS
(54) French Title: PROCEDES D'ELIMINATION DE CELLULES TUMORALES PAR CIBLAGE D'ANTIGENE INTERNE EXPOSE SUR DES CELLULES TUMORALES APOPTOTIQUES
Status: Deemed expired
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 39/395 (2006.01)
  • C07H 21/04 (2006.01)
  • C07K 16/00 (2006.01)
(72) Inventors :
  • EVANS, ELIZABETH E. (United States of America)
  • PARIS, MARK J. (United States of America)
  • SAHASRABUDHE, DEEPAK M. (United States of America)
  • SMITH, ERNEST S. (United States of America)
  • ZAUDERER, MAURICE (United States of America)
(73) Owners :
  • VACCINEX, INC. (United States of America)
(71) Applicants :
  • VACCINEX, INC. (United States of America)
(74) Agent: MBM INTELLECTUAL PROPERTY LAW LLP
(74) Associate agent:
(45) Issued: 2014-02-04
(86) PCT Filing Date: 2004-12-06
(87) Open to Public Inspection: 2005-06-23
Examination requested: 2009-11-30
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2004/040573
(87) International Publication Number: WO2005/055936
(85) National Entry: 2006-06-01

(30) Application Priority Data:
Application No. Country/Territory Date
60/526,572 United States of America 2003-12-04
60/531,688 United States of America 2003-12-23

Abstracts

English Abstract




The invention provides in vitro and in vivo methods of killing cancer cells,
including therapeutic methods in humans, and also provides antibodies specific
for the cancer specific antigen C35, and polynucleotides encoding such
antibodies, as well as therapeutic and diagnostic methods of using such
antibodies.


French Abstract

L'invention concerne des procédés in vitro et in vivo d'élimination de cellules cancéreuses, notamment des procédés thérapeutiques effectués sur des êtres humains, ainsi que des anticorps spécifiques à l'antigène C35 spécifique du cancer et des polynucléotides codant ces anticorps, ainsi que des procédés thérapeutiques et diagnostiques d'utilisation de ces anticorps.

Claims

Note: Claims are shown in the official language in which they were submitted.




200
THE EMBODIMENTS FOR WHICH AN EXCLUSIVE PROPERTY OR
PRIVILEGE IS CLAIMED ARE DEFINED AS FOLLOWS:
1. Use of an apoptosis-inducing therapy and an antibody to treat cancer by
killing
cancer cells that express C35, wherein said antibody is specific for C35 and
wherein said
antibody is for administration at a time such that said antibody binds to C35
expressed on
the surface of said cancer cell at a time when apoptosis has been induced or
is being
induced in said cancer cell, thereby killing cancer cells undergoing
apoptosis.
2. The use according to claim 1, wherein said antibody is conjugated to or
complexed with a toxin.
3. The use according to claim 1 or 2, wherein said antibody is selected
from the
group consisting of:
a) an antibody comprising the VH region amino acid sequence defined in SEQ
ID NO: 8,
b) an antibody comprising the VL region amino acid sequence defined SEQ ID
NO: 10,
c) an antibody comprising the VH region amino acid sequence defined SEQ ID
NO: 4,
d) an antibody comprising the VL region encoded by clone 1F2K amino acid
sequence defined SEQ ID NO: 6,
e) an antibody comprising the VH region of (a) and the VL region of (b),
f) an antibody comprising the VH region of (c) and the VL region of (d),
g) an antibody comprising at least one of CDR1, CDR2 or CDR3 of the VH
region encoded by SEQ ID NO: 56,
h) an antibody comprising at least one of CDR1, CDR2 or CDR3 of the VH
region encoded by SEQ lD NO: 60,
i) an antibody comprising at least one of CDR1, CDR2, or CDR3 of the VL
region encoded by SEQ ID NO: 58,
j) a chimeric antibody comprising the VH region of (a) or (c),
k) a chimeric antibody comprising the VL region of (b) or (d),




201
l) a chimeric antibody comprising the VH region of (a) and the VL region
of (b),
m) a chimeric antibody comprising the VH region of (c) and the VL region of
(d),
n) the chimeric antibody of (j), (k), (l), or (m) which is a human chimeric
antibody,
o) a humanized antibody comprising 1, 2, 3, 4, 5, or 6 CDRs of the antibody of

(e) or (f), and
p) an antibody which binds the epitope bound by the antibody of any one of (a)

to (o).
4. The use according to claim 3, wherein said antibody comprises the
humanized
antibody of (o).
5. The use according to any one of claims 1 to 4, wherein said antibody is
selected
from the group consisting of a whole antibody, an antibody fragment, and a
domain-
deleted antibody, and wherein said antibody is for administration 0-6 hours,
or 6-12
hours, or 6-24 hours, or 6-36 hours, or 6-48 hours, or 6-72 hours, or 6-96
hours, or 6-120
hours, or 12-24 hours, or 12-36 hours, or 12-48 hours, or 12-72 hours, or 12-
96 hours, or
12-120 hours, or 24-36 hours, or 24-48 hours, or 24-72 hours, or 24-96 hours,
or 24-120
hours, or 36-48 hours, or 36-72 hours, or 36-96 hours, or 36-120 hours after
apoptosis
has been induced or is being induced in said cancer cell.
6. The use according to any one of claims 1 to 5, wherein said use is in
vitro.
7. The use according to any one of claims 1 to 5, wherein said use is in
vivo.
8. The use according to any one of claims 1 to 5, wherein said use is for
treatment of
a mammal in need of cancer treatment.
9. The use according to claim 8, wherein said mammal is in need of cancer
treatment
for a C35-associated cancer selected from the group consisting of: breast
cancer, ovarian



202
cancer, bladder cancer, lung cancer, prostate cancer, pancreatic cancer, colon
cancer, and
melanoma.
10. The use according to claim 9, wherein the mammal is a human.
11. The use according to claim 2, wherein the toxin is a radioisotope, a
radionuclide,
a cytotoxin, or a cytotoxic prodrug.
12. The use according to claim 8, wherein said mammal is in need of
eradication of
smaller tumors and/or micrometastases.
13. The use according to claim 12, wherein the mammal is a human.
14. The use according to any one of claims 1 to 13, wherein said antibody
is an
immunoglobulin isotype selected from the group consisting of IgG3, IgM, IgA,
IgD, and
IgE.
15. The use according to any one of claims 1 to 14, wherein said antibody
is a
domain-deleted antibody.
16. The use according to claim 15, wherein said domain-deleted antibody is
a CH2
domain-deleted antibody.
17. The use according to any one of claims 1 to 13, wherein said antibody
is an
antibody fragment selected from the group consisting of an Fab, an F(ab')2, an
scFV, a
minibody, a diabody, a triabody, and a tetrabody.
18. The use according to any one of claims 1 to 17, wherein said use
comprises
multiple uses of the antibody.



203
19. Use of an antibody in the manufacture of a medicament for killing
cancer cells
that expresses C35, wherein said medicament is for use in combination with an
apoptosis-
inducing therapy, wherein said antibody is specific for C35, and wherein said
medicament is for administration at a time such that said antibody binds to
C35 expressed
on the surface of said cancer cell at a time when apoptosis has been induced
or is being
induced in said cancer cell, thereby killing cancer cells undergoing
apoptosis.
20. Use of an apoptosis-inducing therapy and an antibody to prevent
reversal of
apoptotic progression in cancer cells that express C35, wherein said antibody
is specific
for C35, and wherein said antibody is for administration at a time such that
said antibody
binds to C35 expressed on the surface of said cancer cell at a time when
apoptosis has
been induced or is being induced in said cancer cell, thereby preventing
reversal of
apoptotic progression in cancer cells.
21. Use of an antibody in the manufacture of a medicament for preventing
reversal of
apoptotic progression in cancer cells, wherein said medicament is for use in
combination
with an apoptosis-inducing therapy, wherein said antibody is specific for C35,
and
wherein said medicament is for administration at a time such that said
antibody binds to
C35 expressed on the surface of said cancer cell at a time when apoptosis has
been
induced or is being induced in said cancer cell, thereby preventing reversal
of apoptotic
progression in said cancer cell.
22. The use according to any one of claims 19 to 21, wherein said antibody
is
conjugated to or complexed with a toxin.
23. The use according to claim 22, wherein the toxin is a radioisotope, a
radionuclide,
a cytotoxin, or a cytotoxic prodrug.
24. The use according to any one of claims 21 to 23, wherein said antibody
is selected
from the group consisting of:



204
a) an antibody comprising the VH region comprising the amino acid sequence of
SEQ ID NO: 8,
b) an antibody comprising the VL region comprising the amino acid sequence of
SEQ ID NO: 10,
c) an antibody comprising the VH region comprising the amino acid sequence of
SEQ ID NO: 4,
d) an antibody comprising the VL region comprising the amino acid sequence of
SEQ ID NO: 6,
e) an antibody comprising the VH region of (a) and the VL region of (b),
f) an antibody comprising the VH region of (c) and the VL region of (d),
g) an antibody comprising at least one of CDR1, CDR2 or CDR3 of the VH
region encoded by SEQ ID NO: 56,
h) an antibody comprising at least one of CDR1, CDR2 or CDR3 of the VH
region encoded by SEQ 1D NO: 60,
i) an antibody comprising at least one of CDR1, CDR2, or CDR3 of the VL
region encoded by SEQ 1D NO: 58,
j) a chimeric antibody comprising the VH region of (a) or (c),
k) a chimeric antibody comprising the VL region of (b) or (d),
l) a chimeric antibody comprising the VH region of (a) and the VL region of
(b),
m) a chimeric antibody comprising the VH region of (c) and the VL region of
(d),
n) the chimeric antibody of (j), (k), (l), or (m) which is a human chimeric
antibody,
o) a humanized antibody comprising 1, 2, 3, 4, 5, or 6 CDRs of the
antibody of (e) or (f), and
p) an antibody which binds the epitope bound by the antibody of any one of (a)

to (o).
25. The use according to claim 24, wherein said antibody comprises the
humanized
antibody of (o).



205
26. The use according to claim 20, wherein said antibody is selected from
the group
consisting of a whole antibody, an antibody fragment, and a domain-deleted
antibody,
and wherein said antibody is for administration 0-6 hours, or 6-12 hours, or 6-
24 hours,
or 6-36 hours, or 6-48 hours, or 6-72 hours, or 6-96 hours, or 6-120 hours, or
12-24
hours, or 12-36 hours, or 12-48 hours, or 12-72 hours, or 12-96 hours, or 12-
120 hours, or
24-36 hours, or 24-48 hours, or 24-72 hours, or 24-96 hours, or 24-120 hours,
or 36-48
hours, or 36-72 hours, or 36-96 hours, or 36-120 hours after apoptosis has
been induced
or is being induced in said cancer cell.
27. The use according to claim 20, wherein said use is in vivo.
28. The use according to claim 20, wherein said use is in vitro.
29. The use according to claim 20, wherein said use is for treatment of a
mammal in
need of cancer treatment.
30. The use according to claim 29, wherein said mammal is in need of cancer

treatment for a C35-associated cancer selected from the group consisting of:
breast
cancer, ovarian cancer, bladder cancer, lung cancer, prostate cancer,
pancreatic cancer,
colon cancer, and melanoma.
31. The use according to claim 29, wherein said mammal is in need of
eradication of
smaller tumors and/or micrometastases.
32. The use according to any one of claims 29 to 31, wherein the mammal is
a human.
33. The use according to any one of claims 19 to 32, wherein said antibody
is an
immunoglobulin isotype selected from the group consisting of IgG3, IgM, IgA,
IgD, and
IgE.



206
34. The use according to any one of claims 19 to 33, wherein said antibody
is a
domain-deleted antibody.
35. The use according to any one of claims 19 to 33, wherein said domain-
deleted
antibody is a CH2 domain-deleted antibody.
36. The use according to any one of claims 19 to 33, wherein said antibody
is an
antibody fragment selected from the group consisting of an Fab, an F(ab')2, an
scFV, a
minibody, a diabody, a triabody, and a tetrabody.
37. The use according to claim 20, wherein said use comprises multiple uses
of the
antibody.

Description

Note: Descriptions are shown in the official language in which they were submitted.


DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PLUS D'UN TOME.
CECI EST LE TOME 1 DE 2
NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des
Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.
THIS IS VOLUME 1 OF 2
NOTE: For additional volumes please contact the Canadian Patent Office.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
METHODS OF KILLING TUMOR CELLS BY TARGETING INTERNAL
ANTIGENS EXPOSED ON APOPTOTIC TUMOR CELLS
BACKGROUND OF THE INVENTION
[0001] Cell growth is a carefully regulated process which responds to
specific
needs of the body. Occassionally, the intricate, and highly regulated
controls dictating the rules for cellular division break down. When this
occurs, the cell begins to grow and divide independently of its homeostatic
regulation resulting in a condition commonly referred to as cancer. In fact,
cancer is the second leading cause of death among Americans aged 25-44.
[0002] Current therapies for cancer include chemotherapy and radiation
therapy. Chemotherapeutic drugs kill cancer cells mainly by inducing
apoptosis (Fisher, D.E., Cell 78:539-542 (1994); Fung, C.Y., and D.E.
Fisher, J. Clin. Oncol. 13:801-807 (1995); Lowe, S .W., et al., Cell 74:957-
967 (1993)). Radiation therapy kills cancer cells by inducing apoptosis
and by other mechanisms. However, chemotherapy and radiation therapy
do not kill all cells in a given tumor, and cells that survive such treatment
continue to grow. Thus, these treatments are often insufficient for
eradicating an entire tumor. There is therefore a need for improved
therapeutic methods of treating cancer.
[0003] Immunotherapeutic strategies for cancer have also been developed
that
target surface membrane markers differentially expressed in tumor cells
using antibodies (e.g., U.S. Patent number: 5,770,195, "Monoclonal

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
2
[00041 Antibodies to the HER2 Receptor", Filed: May 23, 1995; Issued,
Jun. 23, 1998). Many antigens differentially expressed in tumors are,
however, not exposed on the surface of tumor cells. As a result, such
intracellular antigens are not suitable as targets for antibody-based
therapeutics. Therefore, there is a need for additional targets for
immunotherapeutic methods of treating cancer.
SUMMARY OF THE INVENTION
Methods Of Killing Cancer Cells
[0005] The present invention provides a method of killing cancer cells by
administering an effective amount of an apoptosis-inducing therapy, and
administering an effective amount of an antibody conjugate or antibody
complex that binds a cancer-associated antigen which is expressed
intracellularly in cancer cells, but which becomes exposed on the cell
surface in cancer cells that are undergoing apoptosis. The timing of
administration of the apoptosis-inducing therapy and the antibody
conjugate or antibody complex is planned such that the antibody reaches
the cancer cell at the time that apoptosis is being or has been induced. In a
preferred embodiment, the cancer associated antigen is a prenylated protein
which, although normally expressed intracellulary, become exposed on the
cell surface in tumor cells that are undergoing apoptosis. In another
preferred embodiment, the prenylated protein is the C35 antigen. The
antibody is conjugated to or complexed with a toxin, which insures that the
cell to which the antibody binds will be killed, and/or surrounding cancer
cells that are exposed to the toxin are killed. In one embodiment, the toxin
is a radioisotope.
[00061 In one embodiment, the method involves administering a
chemotherapeutic agent followed by or simultaneous with an antibody or
fragment or variant thereof that is conjugated to a radioactive agent.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
3
[0007] In another embodiment, the method involves administering an antibody
that is not conjugated to or complexed with a toxin, and cells which bind
the antibody die.
[0008] The method of the invention may be performed in vitro or in vivo,
and
may be used as a therapeutic in a patient, including a mammal such as a
human.
Antibodies Against C35 And Methods Of Using C35 Antibodies
[0009] The present Mention also provides antibodies that bind C35
polypeptides. The present invention encompasses antibodies (including
molecules comprising, or alternatively consisting of, antibody fragments or
variants thereof) that immunospecifically bind to a C35 polypeptide or
polypeptide fragment or variant of a C35 polypeptide such as that of SEQ
ID NO:2.
[0010] The present inventors have generated mouse and human antibodies that
immunospecifically bind one or more C35 polypeptides (e.g., SEQ ED
NO:2) and polynucleotides encoding VH and VL regions from these
antibodies. Thus, the invention encompasses these polynucleotides,
inlcuding those set forth in SEQ ID NO:s 56, 58, and 60, and those listed
in Tables 2 and 3 below, which were deposited with the American Type
Culture Collection ("ATCC") on the dates listed in Tables 2 and 3 and
given the ATCC Deposit Numbers identified in Tables 2 and 3. The
ATCC is located at 10801 University Boulevard, Manassas, VA 20110-
2209, USA. The ATCC deposit was made pursuant to the terms of the
Budapest Treaty on the international recognition of the deposit of
microorganisms for purposes of patent procedure.
[0011] The present invention also encompasses the deposited polynucleotide
clones that encode VH and VL regions that inu-nunospecifically bind one
or more C35 polypeptides (e.g., SEQ ID NO:2), cells comprising the
deposited polynucleotides, antibodies comprising VH and/or VL regions

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
4
encoded by the deposited polynucleotides, polynucleotides encoding such
antibodies, and cells comprising such polynucleotides. The present
invention also encompasses cells comprising the polynucleotides of SEQ
ID NO:s 56, 58, and 60, antibodies comprising VH and/or VL regions
encoded by SEQ JD NO:s 56, 58, and 60, polynucleotides encoding such
antibodies, and cells comprising such polynucleotides. Such antibodies
may or may not have the same epitope specificity as the original antibodies
comprising the VH and VL regions encoded by the deposited
polynucleotides, and may or may not have an affinity for C35 the same as
or higher than the affinity of the original antibodies. In one embodiment,
the antibodies of the present invention bind the epitope representing
residues 105 to 115 of the native C35 sequence.
[0012] Further, the present invention encompasses antibodies comprising, or
alternatively consisting of, fragments or variants of these antibodies (e.g.,
scFvs, diabodies, triabodies, tetrabodies, minibodies, heavy chains, VH
regions, VH CDRs (Complementarity Determining Regions), light chains,
VL regions, or VL CDRs) having an amino acid sequence of any one of the
VH, VH CDRs, VLs, VL CDRs encoded by a polynucleotide of the
invention. Such antibodies may or may not have the same epitope
specificity as the original antibodies comprising the VH and VL regions
encoded by the deposited polynucleotides, and may or may not have an
affinity for C35 the same as or higher than the affinity of the original
antibodies.
[0013] The present invention also provides antibodies or fragments or
variants
thereof that bind one or more C35 polypeptides, and which are coupled to
a detectable label, such as an enzyme, a fluorescent label, a luminescent
label, or a bioluminescent label. The present invention also provides
antibodies or fragments or variants thereof that bind one or more C35
polypeptides, and which are coupled to a therapeutic or a toxin, e.g., a
radioactive material. In one embodiment, the antibodies of the present
invention are coupled to a radioisotope.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
[0014] The present invention also provides for a nucleic acid molecule(s),
generally isolated, encoding an antibody (including molecules, such as
scFvs, diabodies, triabodies, tetrabodies, minibodies, VH regions, or VL
regions, that comprise, or alternatively consist of, an antibody fragment or
variant thereof) of the invention. The present invention also provides a
host cell transformed with a nucleic acid molecule encoding an antibody
(including molecules, such as scFvs, diabodies, triabodies, tetrabodies,
minibodies, VH regions, or VL regions, that comprise, or alternatively
consist of, an antibody fragment or variant thereof) of the invention and
progeny thereof. The present invention also provides a method for the
production of an antibody (including a molecule comprising, or
alternatively consisting of, an antibody fragment or variant thereof) of the
invention. The present invention further provides a method of expressing
an antibody (including a molecule comprising, or alternatively consisting
of, an antibody fragment or variant thereof) of the invention from a nucleic
acid molecule.
[0015] The present invention relates to methods and compositions for
treating
cancer comprising administering to a mammal, preferably a human, an
effective amount of one or more antibodies or fragments or variants
thereof, or related molecules, that immunospecifically bind a C35
polypeptide or a fragment or variant thereof. In preferred embodiments, the
present invention relates to antibody-based methods and compositions for
treating breast cancer, ovarian cancer, bladder cancer, lung cancer, prostate
cancer, pancreatic cancer, colon cancer, and melanoma.
[0016] In a preferred embodiment, the present invention relates to a
combination therapy for treating cancer comprising administering to a
mammal, preferably a human, an effective amount of a chemotherapeutic
and an effective amount of one or more antibodies, or fragments or
variants thereof, that are conjugated with a toxin, e.g., a radioactive
material.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
6
[0017] The present invention also encompasses methods and compositions for
detecting, diagnosing, or proposing cancer comprising administering to a
mammal, preferably a human, an effective amount of one or more
antibodies or fragments or variants thereof, or related molecules, that
immunospecifically bind to C35 or a fragment or variant thereof. In
preferred embodiments, the present invention relates to antibody-based
methods and compositions for detecting, diagnosing, or prognosing breast
cancer, ovarian cancer, bladder cancer, lung cancer, prostate cancer,
pancreatic cancer, colon cancer, and melanoma.
[0018] Another embodiment of the present invention includes the use of the
antibodies of the invention as a diagnostic tool to monitor the expression
of C35 or in cancer. in certain embodiments, the method may also be
employed as a diagnostic to confirm the efficacy of an apoptosis inducing
regimen.
[0019] These and other aspects of the invention are described in further
detail
below.
BRIEF DESCRIPTION OF THE FIGURES
[0020] Figure 1 shows C35 surface staining of breast tumor cells following
radiation induced apoptosis in 21MT1 breast tumor cells that express the
C35 tumor antigen. Fig. 1A shows that untreated live cells (PI negative),
that are not undergoing apoptosis (Annexin V negative) do not express
C35 on the surface membrane as evidenced by absence of differential
staining with anti-C35 antibody and the isotype control antibody. Fig. 1B
shows, similarly, that irradiated tumor cells that remain viable (PI
negative) and have not been induced to undergo apoptosis (Annexin V
negative) also do not express C35 on the tumor cell surface membrane.
Fig. IC shows, in contrast, that irradiated tumor cells that are viable (PI
negative), but undergoing apoptosis (Annexin V positive), are clearly

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
7
differentially stained with anti-C35 antibodies as compared to isotype
control antibody.
[0021] Figure 2 shows C35 surface staining of breast tumor cells following
mitomycin C drug induced apoptosis. Fig. 2A shows that untreated live
cells (PI negative), that are not undergoing apoptosis (Annexin V
negative), do not express C35 on the surface membrane as evidenced by
absence of differential staining with anti-C35 antibody and the isotype
control antibody. Fig. 2B shows, similarly, that mitomycin C treated tumor
cells that remain viable (PI negative) and have not been induced to undergo
apoptosis (Annexin V negative) also do not express C35 on the tumor cell
surface membrane. Fig. 2C shows, in contrast, that mitomycin C treated
tumor cells that are viable (PI negative), but undergoing apoptosis
(Annexin V positive), are clearly differentially stained with anti-C35
antibodies as compared to isotype control antibody.
[0022] Figures 3A-3C show that TaxolTm induces apoptosis, resulting in
exposure of C35 on the surface of apoptotic tumor cells. 24 hours
following treatment with 0.5 uM Taxolmi, 21MT1 tumor cells were stained
with annexin V-FITC, propidium iodide, and with either 100 ng anti-C35
antibody 1F2 (dark line) or isotype control (grey fill) antibody. Both
antibodies were directly conjugated to Alexa-647. Histograms were gated
on the cells undergoing apoptosis (annexinV positive/PI negative).
Antibodies were pre-incubated with PAB buffer (Fig. 3A), 100-fold molar
excess recombinant C35 protein (Fig. 3B), or 100-fold molar excess 13-
galactosidase protein (Fig. 3C).
[0023] Figure 4 shows that anti-C35 monoclonal antibody localizes to
necrotic
regions of a C35+ tumor. BALB/c mice were engrafted on opposite flanks
with syngeneic non-small cell lung cancer derived Line 1 tumor cells that
either had or had not been tran.sfected with human C35. C35 protein
expression was confirmed by immunohistochemical staining with anti-C35
antibodies. After 14 days in vivo growth, animals received intravenous

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
8
injection of 1251-labeled anti-C35 antibody. Animals were sacrificed 120
hrs after injection of radiolabeled antibodies and the concentration of anti-
C35 antibodies in C35-positive and C35-negative tumors was determined
by exposure of a tumor section to film. Figure 4A shows that radiolabeled
anti-C35 antibodies concentrate only in the C35-positive and not the C35-
negative tumors. Figures 4B and 4C compare the distribution of label and
an H&E stain for intact cells within the tumors, confirming that under
these conditions the labeled anti-C35 antibodies concentrated specifically
in the necrotic regions of the C35-positive tumor.
[0024] Figure 5 shows the effect on tumor volume of combination
radioimmunotherapy with 131I-labeled 1B3 anti-C35 murine monoclonal
antibody and chemotherapy (fluorouracil, 150 mg/kg; leucovorin, 100
mg/kg) in Swiss nude mice grafted with Colau.C35 tumor cells.
Chemotherapy was initiated on day 11 after tumor graft and 300 fiCi of
1311-labeled 1B3 anti-C35 antibody was administered on day 14. Tumor
growth was followed for up to 8 weeks.
[0025] Figure 6 shows the effects on tumor volume of the combined modality
treatment of chemotherapy and radioimmunotherapy. Swiss nude mice
were grafted with Colau.C35 cells on day 0. Chemotherapy: Cisplatin
administered at 2 mg/kg i.v. on days 15 & 18; 5FU/LV administered at
180/120 mg/kg i.v. on day 18. Radioimmunotherapy: 300 piCi (-50 ptg) of
1311-labeled murine 1B3 anti-C35 IgG was administered on day 21.
[0026] Figure 7 shows equivalent expression in naturally-expressing and C35-

transfected human breast and colon tumors. Cells were stained with Alexa-
647 conjugated anti-C35 MAb 1F2 or isotype control. MFI X is the ratio of
the mean fluorescence intensity of 1F2/mean fluorescence intensity of
isotype control. H16N2, derived from normal breast epithelium, and
MDAMB231, a breast tumor, and Colau, a colon tumor, express low basal
levels of C35. 21MT1, derived from breast carcinoma, naturally expresses
high levels of C35. Colau and MDA231 were transduced with empty

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
9
vector (null) or human C35 recombinant vector. All tumors were grown in
vivo, tumors were excised, dissociated and stained.
[0027] Figure 8 shows toxicity of chemotherapy, radioimmunotherapy, and
combination therapy in Swiss nude mice as determined by weight loss.
[0028] Figure 9 shows the expected peptide fragments following complete
digestion of 6x His-tagged recombinant human C35 (rhC35) with Lys-C
endoprotease. The full sequence of rhC35, including the amino terminal
6x His tag addition is shown. Amino acid poistions are numbered relative
to the amino terminal methionine (bold M) of the native human C35
sequence. The asterisks by the first and third lysine (K) residues indicate
that digestion at these positions is inefficient, and some longer fragments
may be generated.
[0029] Figure 10 shows a comparison of 1B3 (Mab 11) or 1F2 and anti-6x His
tag staining of Western blots indicating the fragment of C35 to which each
antibody binds.
[0030] Figure 11 shows that MAb 165 is C35-specific. 141D10 recombinant
vaccinia virus was co-infected into HeLa cells with UII8 recombinant
vaccinia virus. The resulting secreted antibody was tested for binding to
C35 or control protein A27L (vaccinia virus protein) by ELISA.
DETAILED DESCRIPTION OF THE INVENTION
OVERVIEW
[0031] A number of studies have described alterations in the surface
membrane of cells undergoing apoptosis. Prominent among these changes
is the early loss of phospholipid asymmetry as reflected in the exposure of
phosphatidylserine on the outer leaflet of the surface membrane. It has
been reported that this alteration in surface membrane composition
facilitates recognition and removal of apoptotic cells by macrophages
(Fadok, V.A., et al., J. ItninunoL /48:2207-2216 (1992)). A general

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
method has been developed that allows detection of cells undergoing
apoptosis by binding of the anticoagulant Annexin V to the exposed
phosphatidylserine molecules (Koopman, G., et al., Blood 84:1415-1420
(1994)).
[0032] Of more general interest is the possibility that expression and
exposure
of other surface membrane molecules, in particular proteins, may be
altered in apoptotic cells. A number of reports have described apoptosis
specific proteins (Grand, R.J.A., et al., Exp. Cell Res. 2/8:439-451 (1995);
U.S. Patent number: 5,972,622, "Method of Detecting Apoptosis Using an
Anti-Human GP46 Monoclonal Antibody", Filed: Feb. 6, 1997; Issued,
Oct. 26, 1999) that appear to be expressed intracellularly. Of more direct
relevance is a report of a monoclonal antibody that detects a 38 kD protein
antigen that becomes associated with the surface membrane and
mitochondrial membranes of apoptotic cells but is undetectable in normal
cells (U.S. Patent number: 5,935,801, "Monoclonal Antibody that Detects
Apoptotic Antigen", Filed: Mar. 29, 1996; Issued, Aug. 10, 1999). Other
antigens have been described that become differentially exposed on or near
the surface of apoptotic keratinocytes (Casciola-Rosen, L.A., et al., J. Exp.
Med. 179:1317-1330 (1994)), and in cells undergoing apoptosis during
embryonic development (Rotello, R.J., et al., Development 120:1421-1431
(1994)). Three defined protein antigens, CD3, CD69 and CD25 have been
shown to be upregulated on the surface membrane of apoptotic thymocytes
(Kishimoto, H., et al., Ji Exp. Med. 181:649-655 (1995)). In each instance
these are surface markers of apoptosis in normal cells and tissues.
Although the same markers might also be associated with tumor cells
undergoing apoptosis, they do not allow apoptotic tumor cells to be
distinguished from normal cells undergoing apoptosis as part of normal
tissue turnover. Therefore, they would not be useful as targets for treating
cancer.
[0033] The present inventors have determined that there is a subset of
intracellular tumor-specific or tumor-associated antigens that become

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
11
exposed on the tumor cell membrane under conditions of chemotherapy or
radiation induced apoptosis and could be effective targets for concentrating
antibody conjugated radioisotopes or toxins within the tumor. Methods
using antibodies against such antigens would be particularly effective
because they could enhance the therapeutic benefits of standard apoptosis-
inducing chemotherapy and radiation therapy in treating cancer. The
present invention identifies tumor-specific antigens that are associated with
internal cell membranes -- in particular, differentially expressed molecules
such as the C35 cancer-specific antigen that express a prenylation motif --
as a class of intracellular tumor antigens that become exposed on the
surface membrane of tumor cells that have been induced to undergo
apoptosis by radiation and/or chemotherapy.
[0034] The present invention describes a method that acts in conjunction
with
the induction of apoptosis (preferably large scale apoptosis) by
chemotherapy or radiation therapy to enhance the eradication of tumors. It
is based on the novel observation that a class of intracellular markers
differentially expressed in tumor cells becomes exposed on the surface of
apoptotic cells where it can be targeted by specific antibodies conjugated
to a toxic payload. The benefits of this method of treatment are several-
fold. For example, this method permits delivery to the tumor environment
of a toxic payload that can destroy other non-apoptotic tumor cells in the
vicinity of the apoptotic target. Additionally, this method can prevent
otherwise viable cells that have initiated the apoptotic process, as
evidenced by alterations in surface membrane constituents, from reversing
the apoptotic progression and resuming growth (Hammill, A.K., et al.,
Exp. Cell Res. 251:16-21 (1999)).
[0035] The present invention targeting apoptotic cells should be
distinguished
from prior inventions targeting necrotic cells (U.S. Patent number:
6,071,491, "Detection of Necrotic Malignant Tissue and Associated
Therapy", Filed: Aug. 9, 1999; Issued, Jun. 6, 2000). Necrosis results in
release of intracellular contents into the extracellular tumor environment.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
12
Some of these intracellular antigens accumulate in that environment and
could be targeted by specific antibodies. However, necrosis is associated
with hypoxic regions of larger tumors that, because of the absence of
oxygen radicals, are relatively resistant to radiation therapy and possibly
radio-immunotherapy. Although there may be some increase in necrosis
following treatment with chemotherapeutic agents (Desrues B., et al., Br. J.
Cancer 72:1076-82, (1995)), the primary action of chemotherapeutic
agents is to increase apoptosis. Therefore, necrosis is a less suitable target

than apoptosis for immunotherapy of cancer and, in particular, eradication
of smaller tumors and micrometastases that are responsible for tumor
spread. Thus, methods that are effective at eradicating small tumors and
micrometastases are especially useful for treating aggressive cancers.
[0036] The present invention should also be distinguished from the
disclosure
in patent application publication number US 2002/0052308 Al (May 2,
2002), which discloses 842 cancer antigens, including an antigen (SEQ ID
NO:966) with a large region identical to a portion of C35 (SEQ ID NO:2).
US 2002/0052308 Al generically discloses the administration of
antibodies against the 842 cancer antigens "alone or in combination with
other types of treatments (e.g., radiation therapy, chemotherapy, hormonal
therapy, immunotherapy and anti-tumor agents)", page 205, paragraph
[0229]. However, the published application does not specify that to be
effective against a C35 related target, C35-specific antibodies conjugated
to a toxin should be administered after apoptosis has been induced in
tumor cells by administration of an apoptosis inducing agent such as
chemotherapy, radiation therapy, or other anti-tumor agents. Indeed,
multiple studies of combination chemotherapy and radioimmunotherapy
directed at antigens that, in contrast to C35, are naturally expressed on the
tumor cell surface membrane have concluded that optimal results are
obtained by administration of the radioimmunotherapeutic antibody prior
to chemotherapy, that is, before apoptosis has been induced (DeNardo S.J.,
et al. Anticancer Res. 18:4011-18, (1998); Clarke K., et al., Clin. Cancer

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
13
Res. 6:3621-28, (2000); Burke P.A., Cancer 94:1320-31(2002); Stein, R. et
al., Cancer 94:51-61 (2002); Odonnel R.T., et al., Prostate 50:27-37
(2002)). The discovery that apoptosis results in surface membrane
exposure of a class of intracellular antigens including C35, which are
prenylated and associated with internal membranes of untreated tumor
cells, distinguishes the present invention. This invention teaches that for
optimal effect, radioimmunotherapy directed at this class of target
molecules is best administered such that the antibodies accumulate at the
tumor site at approximately the same time that apoptosis has been induced
in tumor cells by administration of an apoptosis inducing agent, or shortly
thereafter. US 2002/0052308 Al does not describe the subcellular location
of the C35 related cancer antigen, nor does it describe how antibodies to
this antigen should be administered for therapeutic effect.
Methods Of Killing Cancer Cells
[0037] The present invention is directed to a method of killing cancer
cells
(also referred to herein as tumor cells) by first administering an effective
amount of an apoptosis-inducing therapy (e.g., a chemotherapeutic agent
and/or radiation), and subsequently administering an effective amount of
an antibody conjugate or antibody complex that binds a cancer-associated
antigen which is expressed intracellularly in tumor cells, but which
becomes exposed on the cell surface in tumor cells that are undergoing
apoptosis. The antibody is conjugated to or complexed with a toxin, as
described below. The toxin insures that the cell to which the antibody
binds will be killed, and/or kills surrounding cells that are also exposed to
the toxin.
[0038] In one embodiment, the method involves the combined use of
chemotherapy and radioimmunotherapy. Prior to the present invention,
combination chemotherapy and radioimmunotherapy posed a problem due to
cumulative dose-limiting bone marrow toxicity. The present invention

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
14
provides for administration of such combination therapy in that chemotherapy
results in exposure of an intracellular antigen to antibodies. Optimal timing
of
administration can be precisely determined employing the methods described
herein and in the Examples.
[0039] In another embodiment, the method involves administering an antibody
that is not conjugated to or complexed with a toxin, and cells which bind
the antibody are killed. In this embodiment, a toxin is not needed to be
conjugated to or complexed with the antibody in order to kill the cells that
bind the antibody. Binding of the antibody itself kills the cell or insures
that it dies. In this embodiment, apoptosis is preferably induced in most or
nearly all of the cancer cells, e.g., most or nearly all of the cells of a
tumor
or metastasis.
[0040] The timing for administering the antibody, antibody conjugate or
antibody complex after the apoptosis-inducing therapy can vary, however
it must be within a certain window of time during which the tumor cells
are undergoing apoptosis. Thus, the antibody, conjugated antibody or
complexed antibody is administered within the time period during which
apoptosis is being induced or is ongoing in the tumor cells that have been
treated with the apoptosis-inducing therapy (e.g., chemotherapeutic agent
and/or radiation). Chemotherapeutic agents generally induce apoptosis 24-
72 hours after administration. Antibodies (e.g., complexed and conjugated
antibodies) against cancer-associated antigens generally accumulate at the
site of a tumor(s) 24-48 hours after administration. Whole antibodies
generally take longer to accumulate than antibody fragments. Therefore, in
general, antibodies (e.g., complexed and conjugated antibodies) against
cancer-associated intracellular antigens should be administered 0-48 hours
after administration of a chemotherapeutic agent (for antibody fragments),
and from 24 hours before, to 24 hours after, administration of a
chemotherapeutic agent (for whole antibodies) in the method of the
invention. In a preferred embodiment, the antibody, or conjugated or
complexed antibody is administered 0-6 hours, or 6-12 hours, or 6-24

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
hours, or 6-36 hours, or 6-48 hours, or 6-72 hours, or 6-96 hours, or 6-120
hours, or 12-24 hours, or 12-36 hours, or 12-48 hours, or 12-72 hours, or
12-96 hours, or 12-120 hours, or 24-36 hours, or 24-48 hours, or 24-72
hours, or 24-96 hours, or 24-120 hours, or 36-48 hours, or 36-72 hours, or
36-96 hours, or 36-120 hours or 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13,
14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32,
33,
34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52,
53,
54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72,
73,
74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92,
93,
94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109,
110, 111, 112, 113, 114, 115, 116, 117, 118, 119, or 120 hours after the
apoptosis-inducing therapy is administered. In another embodiment, the
antibody (e.g., complexed and conjugated antibodies) is administered prior
to the apoptosis-inducing therapy, for example, 0-6 hours, 0-12, 0-24, 6-
12, 6-24, 12-24, 24, 23, 22, 21, 20, 19, 18, 17, 16, 15, 14, 13, 12, 10, 9, 8,

7, 6, 5, 4, 3, 2 hours, or 1 hour before the apoptosis-inducing therapy is
administered. In another embodiment, the antibody (e.g., complexed and
conjugated antibodies) is administered simultaneously with the apoptosis-
inducing therapy.
[0041] Apoptosis-inducing therapies useful in the method of the invention
are
discussed below. By "tumor" or "cancer" or "hyperproliferative disease"
is meant all neoplastic cell growth and proliferation, whether malignant or
benign, incuding all transformed cells and tissues and all cancerous cells
and tissues.
[0042] Examples of cancer include, but are not limited to, carcinoma,
lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
More particular examples of such cancers include squamous cell cancer
(e.g. epithelial squamous cell cancer), lung cancer including small-cell
lung cancer, non-small cell lung cancer, adenocarcinoma of the lung and
squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular
cancer, gastric or stomach cancer including gastrointestinal cancer,

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
16
pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver
cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal
cancer, colorectal cancer, endometrial cancer or uterine carcinoma, salivary
gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer,
thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as
well as head and neck cancer. Other examples of cancer are listed under
"Hyperproliferative Diseases," below.
[0043] By "surrounding" cancer cells is meant cancer cell(s) sufficiently
close
as to be killed by the toxin conjugated to or complexed with the antibody.
[0044] The method of the invention may be performed in vitro or in vivo,
and
may be used as a therapeutic in a patient, including a mammal such as a
human.
Cancer-Associated, Intracellular Antigens
[0045] The method of the invention is directed to the administration of
antibodies (e.g., complexed and conjugated antibodies) against cancer-
associated, intracellular antigens at a time such that the antibodies
accumulate at the cancer site during or after apoptosis has been induced by
the administration of an apoptosis-inducing therapy.
[0046] The terms "antigen" and "epitope" are well understood in the art and
refer to the portion of a macromolecule which is specifically recognized by
a component of the immune system, e.g., an antibody or a T-cell antigen
receptor. The term epitope includes any protein determinant capable of
specific binding to an immunoglobulin. Epitopic determinants usually
consist of chemically active surface groupings of molecules such as amino
acids or sugar side chains and usually have specific three dimensional
structural characteristics, as well as specific charge characteristics.
[0047] By "cancer-associated antigen" or "tumor-associated antigen" is
meant
an antigen that is expressed preferentially by cancer cells relative to non-
cancerous cells of the same cell type or non-cancerous cells from the same

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
17
tissue. Cancer-associated antigens may not be exclusively expressed by
cancer cells (i.e., other normal cells may still express these antigens).
However, the expression of cancer-associated antigens is generally
consistently upregulated in cancers of a particular type or types. Cancer-
associated antigens are antigens, preferably proteins, that may elicit
specific immune responses in animals having particular types of cancer and
thus, include cancer-associated antigens and fragments of cancer-
associated antigens. The terms include not only complete tumor-associated
antigens, but also epitope-comprising portions (fragments) thereof. A
tumor-associated antigen (TAA) may be one found in nature, or may be a
synthetic version of a TAA found in nature, or may be a variant of a
naturally-occurring TAA, e.g., a variant that has enhanced immunogenic
properties.
[0048] Many cancer-associated antigens are known in the art, and routine
methods for determining whether a newly identified gene or protein is
associated with a type of cancer or tumor are known. Such methods
include northern blot analysis, differential display, SAGE, two dimensional
protein gel electrophoresis or tandem mass spectrometry, Southern blot
analysis, and other methods to detect an increased level of mRNA or
protein expression or specific gene amplification in cancer cells from a
particular type of cancer or tumor, in comparison with normal (non-
cancerous or non-transformed) cells from the tissue of origin of the cancer
or tumor, or in comparison to normal cells from other tissues. Other
methods include analysis by tandem mass spectrometry of peptides
differentially expressed in association with MHC molecules of tumor vs.
normal cells.
[0049] By an "intracellular" cancer-associated antigen is meant a cancer-
associated antigen that is expressed on an intracellular membrane of a
tumor cell (a cancerous or transformed cell) that is not undergoing
apoptosis. Internal membranes on which "intracellular" cancer-associated
antigens may be expressed include the endoplasmic reticulum (ER), other

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
18
cytoplasmic membrane bound vesicles including endosomal and lysosomal
vesicles, mitochondrial membranes, and the nuclear membrane Preferred
"internally expressed" cancer-associated antigens include prenylated
proteins expressed on the endoplasmic reticulum and/or membranes of
endosomal or lysosomal vesicles. Examples of prenylated cancer-
associated proteins are listed in Table 1. Prenylated cancer-associated
proteins may also exclude C35 or any individual protein listed in Table 1,
or any combination thereof. For example, prenylated cancer-associated
proteins may exclude CENP-F kinetochore protein, CAAX box protein 1,
DnaJ homolog subfamily A member 1 or 2, or Guanine nucleotide-binding
protein G(I)/G(S)/G(0) gamma-5 subunit.
[0050] For a newly identified cancer-associated antigen, e.g., a cancer-
associated protein, routine methods to determine whether it is prenylated
include searching for a string of nucleotides at the 3' end of the
gene/protein sequence that corresponds to a prenylation motif. A number
of eukaryotic proteins are post-translationally modified by the attachment
of either a famesyl or a geranyl-geranyl group to a Cysteine residue
(Glomset, J.A., et al., Trends Biochem. Sci. 15:139-142 (1990); Lowy,
D.R., and Willumsen, B.M., Nature 341:384-385 (1989); Imagee, A.I.,
Biochem. Soc. Trans. /7:875-876 (1989); Powers, S., Curr. Biol.
1:114-
116 (1991)). The modification occurs on cysteine residues that are three
residues away from the C-terminal extremity; the two residues that
separate this cysteine from the C-terminal residue are generally aliphatic
(Ali). This Cys-Ali-Ali-X pattern is generally known as the CAAX box.
Aliphatic amino acids include isoleucine, valine, leucine, alanine, and
proline. The last four amino acids of C35 are CVIL. The leucine at the
terminal position indicates that this is a substrate for the prenyltransferase

GGTase I which results in addition of a geranyl-geranyl group (Moomaw
and Casey, J. Biol. Chem. 267, 17438-17443 (1992)).
[0051] Thus, the method of the invention also provides for determining
whether a cancer-associated protein is an intracellular protein which

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
19
becomes exposed at the cell surface in cells undergoing apoptosis, prior to
administering the antibody (e.g., complexed and conjugated antibodies) or
apoptosis-inducing agent. Such a determination may encompass analyzing
the amino acid sequence of a candidate protein for the CAAX box at its C-
terminus by computer or manually, and/or performing assays to determine
whether the candidate protein is expressed extracellularly after induction of
apoptosis in cells expressing that protein. Such assays are know in the art
and described herein (see, e.g. Examples 1 and 2) and include inducing
apoptosis in cells expressing a candidate protein and using antibodies (e.g.,
labelled antibodies) specific for the candidate protein to detect the protein
on the cell surface of the apoptotic cells.
Table 1. Genes Encoding Cancer-Associated Prenylated Proteins
Gene Name GenBank Tumor Types Protein Name
Accession No.
CENPF P49454 Colon, kidney,liver, CENP-F kinetochore protein
nervous, skin (Centromere protein F)
(Mitosin) (AR antigen)
CXX1 015255 Breast, pancreas CAAX box protein 1 (Cerebral
protein-5) (hucep-5)
DNAJA1 OR P31689 GI tract, pancreas, DnaJ homolog subfamily A
1-ISJ2 OR stomach, prostate member 1 (Heat shock 40 kDa
HSPF4 OR protein 4) (DnaJ protein
DNAJ2 OR homolog 2) (HSJ-2) (HSDJ)
HDJ2
DNAJA2 OR 060884 Colon, stomach DnaJ homolog subfamily A
HIRIP4 member 2 (H1RA interacting
protein 4) (Cell cycle
progression restoration gene 3
protein) (Dnj3)
GNG5 OR P30670 Brain Guanine nucleotide-binding
GNGT5 protein G(I)/G(S)/G(0) gamma-
subunit
GNG10 OR P50151 - Genitourinary, breast Guanine nucleotide-binding
GNGT10 protein G(1)/G(S)/G(0) gamma-
subunit
GNG12 Q9UB16 - Colon, cartilage, Guanine nucleotide-binding
genitourinary protein G(I)/G(S)/G(0) gamma-
12 subunit
LMNB1 OR P20700 - Colon, genitourinary, Lamin B1

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
Gene Name GenBank Tumor Types Protein Name
Accession No.
LMN2 OR breast
LMNB
LMNB2 OR Q03252 Brain, colon, breast, Lamin B2
LMN2 prostate, stomach, uterus
LMNA OR P02545 Pancreas, skin, stomach, Lamin A/C (70 kDa lamin)
LMN1 uterus
PPP1R16A Q96134 Stomach Protein phosphatase 1
regulatory inhibitor subunit
16A
PXF OR HK33 P40855 Ovary Peroxisomal farnesylated
OR PEX19 protein (33 kDa housekeeping
protein) (Peroxin 19)
RAB11B OR Q15907 Prostate, skin, uterus, Ras-related protein Rab-
11B
YPT3 kidney (GTP-binding protein YPT3)
RAB22A Q9UL26 Hepatocellular Ras-related protein Rab-22A
(Rab-22)
RAB7 P51149 Colon, head and neck, Ras-related protein Rab-7
stomach
MEL OR P24407 Nervous, ovary, pancreas, Ras-related protein Rab-8
(Rab-
RAB8 lung, uterus 8A) (Oncogene c-mel).
RAC1 P15154 Ovarian, colon Ras-related C3 botulinum toxin
substrate 1 (p21-Racl) (Ras-
like protein TC25)
RAC2 P15153 Kidney, liver, lung, Ras-related C3 botulinum
toxin
nervous substrate 2 (p21-Rac2) (Small
G protein) (GX)
RAC3 014658 Breast, prostate Ras-related C3 botulinum toxin
substrate 3 (p21-Rac3)
RAP2A P10114 Prostate Ras-related protein Rap-2a
RAP 1B P09526 Skin Ras-related protein Rap-1b
(GTP-binding protein smg
p21B)
HRAS OR P01112 Bladder, thyroid Transforming protein p21/H-
HRAS1 Ras-1 (c-H-ras)
KRAS2 OR P01116 Colon, ovary Transforming protein p21A (K-
RASK2 Ras 2A) (Ki-Ras) (c-K-ras).
NRAS P01111 Myeloma, melanoma, Transforming protein N-Ras
leukemia
RAB10 088386 Colon, pancreas Ras-related protein Rab-10
RAB13 P51153 Brain, liver, breast Ras-related protein Rab-13
RABlA OR P11476 Colon, ovary, pancreas Ras-related protein Rab-1A
RAB1 (YPT1-related protein).
RAB1B Q9110U4 Breast, prostate, pancreas Ras-related protein Rab-
1B
RAB25 OR P57735 Head and neck, ovary, Ras-related protein Rab-25
CATX8 pancreas (CATX-8).
RAB27A OR P51159 Breast Ras-related protein Rab-27A
RAB27 (Rab-27) (GTP-binding protein

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
21
Gene Name GenBank Tumor Types Protein Name
Accession No.
Ram).
RAB30 Q15771 Ovary, lung Ras-related protein Rab-30
RAB31 OR Q13636 Pancreas Ras-related protein Rab-31
RAB22B (Rab-22B).
RAB32 Q13637 Brain Ras-related protein Rab-32
RAB35 OR Q15286 Prostate Ras-related protein Rab-35
RABIC OR (Rab-1C) (GTP-binding protein
RAY RAY).
RAB36 095755 Nasopharyngeal Ras-related protein Rab-36
RAB38 P57729 Melanoma Ras-related protein Rab-38
(Antigen NY-MEL-1).
RAB3A P20336 Insulinoma Ras-related protein Rab-3A
RAB3D OR 095716 Ovary, prostate, breast Ras-related protein Rab-
3D
RAB16
RAB4A OR P20338 Stomach Ras-related protein Rab-4A
RAB4
RAB5C P51148 Skin, breast Ras-related protein Rab-5C
(RAB5L) (L1880).
RAB6A OR P20340 Colon Ras-related protein Rab-6A
RAB6 (Rab-6).
ARHA OR P06749 Breast Transforming protein RhoA
ARH12 OR (H12).
RHOA OR
RH012
ARHB OR P01121 Breast Transforming protein RhoB
ARH6 OR (H6).
RHOB
ARHC OR P08134 Pancreas Transforming protein RhoC
ARH9 OR (H9).
RHOC
ARHD OR 000212 Breast, pancreas, stomach Rho-related GTP-binding
RHOD protein RhoD (Rho-related
protein HP1) (RhoHP1).
ARHG OR P35238 Lymphoreticular, Rho-related GTP-binding
RHOG pancreas, skin protein RhoG (Sid10750).
ARM OR Q15669 AIDS-associated NHL, Rho-related GTP-binding
TTF NHL, primary lymphoma protein RhoH (GTP-binding
protein TTF).
RRAS2 OR P17082 Ovary, breast Ras-related protein R-Ras2
TC21 (Ras-like protein TC21)
(Teratocarcinoma oncogene).
RRAS P10301 Pancreas Ras-related protein R-Ras
(p23).
[0052] In the various embodiments of this invention, prenylated proteins
are
treated as a single class with closely related properties relative to

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
22
translocation to the surface exposed membrane of cells undergoing
apoptosis.
Method Of Treatment
[0053] The method described above for killing cancer cells can be used in
vivo, as a method of treating a mammalian subject.
[0054] By "subject" or "individual" or "patient" or "mammal," which terms
are used interchangeably herein, is meant any subject, particularly a
mammalian subject, for whom diagnosis or therapy is desired.
Mammalian subjects include humans, domestic animals, farm animals, and
zoo, sports, or pet animals such as dogs, cats, guinea pigs, rabbits, rats,
mice, horses, cattle, cows, and so on.
[0055] The term "treat" or "treatment" refer to both therapeutic treatment
and
prophylactic or preventative measures, wherein the object is to prevent or
slow down (lessen) an undesired physiological change or disorder, such as
the development or spread of cancer. Beneficial or desired clinical results
include, but are not limited to, alleviation of symptoms, diminishment of
extent of disease, stabilized (i.e., not worsening) state of disease, delay or

slowing of disease progression, amelioration or palliation of the disease
state, and remission (whether partial or total), whether detectable or
undetectable. "Treatment" can also mean prolonging survival as compared
to expected survival if not receiving treatment. Those in need of treatment
include those already with the condition or disorder as well as those prone
to have the condition or disorder or .those in which the condition or
disorder is to be prevented. Any of these treatment types or types of
patients may also be excluded.
[0056] Preferred in the method of the invention are the combinations shown
in
Table 1, in which an antibody (e.g., complexed and conjugated antibodies)
specific for a listed prenylated protein, is used to treat a cancer type
listed
for that prenylated protein. For example, an antibody specific for CENP-F

CA 02548180 2011-12-28
23
kinetochore may be used in the method of the invention to treat colon,
kidney, liver, nervous, and skin cancers; an antibody specific for CAAX
box protein 1 may be used to treat breast and pancreatic cancers; an
antibody specific for DNAJ homolog subfamily A member 1 may be used
to treat gastrointestinal tract, pancreatic, stomach, and prostate cancers,
and
so on. In addition, antibodies specific for C35 protein may be used to treat,
diagnose, or detect, breast cancer, ovarian cancer, bladder cancer, lung
cancer, prostate cancer, pancreatic cancer, colon cancer, and melanoma.
[0057] Also preferred in the method of the invention are a combination
chemotherapy/radioimmunotherapy for treating cancer comprising
administering to a mammal, preferably a human, an effective amount of a
chemotherapeutic, followed by or simultaneous with an antibody of the
invention that is conjugated to a toxin, preferably a radioactive material.
[0058] In another embodiment, an antibody of the invention that is
conjugated
with a toxin, preferably a radioactive material, is administered alone.
Antibodies
[0059] As used herein, the
term "antibodies" or "immunoglobulins" refers to
antibodies comprised of two immunoglobulin heavy chains and two
immunoglobulin light chains as well as a variety of forms besides
antibodies; including, for example, Fv, Fab, and F(ab')2 as well as
bifunctional hybrid antibodies (e.g., Lanzavecchia et al., Eur. J. Immunol.
17, 105 (1987)) and single chains (e.g., Huston et al., Proc. Natl. Acad.
Sci. U.S.A., 85, 5879-5883 (1988) and Bird et al., Science 242, 423-426
(1988)). (See, generally,
Hood
et al., Immunology, Benjamin, N.Y., 2ND ed. (1984), Harlow and Lane,
Antibodies. A Laboratory Manual, Cold Spring Harbor Laboratory (1988)
and Hunkapiller and Hood, Nature, 323, 15-16 (1986)).

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
24
[0060] Antibodies of the invention include, but are not limited to,
polyclonal,
monoclonal, multispecific, human, humanized or chimeric antibodies,
single chain antibodies, Fab fragments, F(a1352 fragments, fragments
produced by a Fab expression library, domain-deleted antibodies
(including, e.g., CH2 domain-deleted antibodies), anti-idiotypic (anti-Id)
antibodies (including, e.g., anti-Id antibodies to antibodies of the
invention), and epitope-binding fragments of any of the above. The term
"antibody," as used herein, refers to immunoglobulin molecules and
immunologically active portions of immunoglobulin molecules, i.e.,
molecules that contain an antigen binding site that immunospecifically
binds an antigen. The immunoglobulin molecules of the invention can be
of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2,
IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.
[0061] Antibodies of the invention also include, but are not limited to,
engineered forms of antibodies and antibody fragments such as diabodies,
triabodies, tetrabodies, and higher multimers of scFvs, as well as
minibodies, such as two scFv fragments joined by two constant (C)
domains. See, e.g., Hudson, P.J. and Couriau, C., Nature Med. 9: 129-134
(2003); U.S. Publication No. 20030148409; U.S. Patent No. 5,837,242.
[0062] The antibodies of the invention may be from any animal origin
including birds and mammals. Preferably, the antibodies are human,
murine (e.g., mouse and rat), donkey, ship rabbit, goat, guinea pig, camel,
horse, or chicken. As used herein, "human" antibodies include antibodies
having the amino acid sequence of a human immunoglobulin and include
antibodies isolated from human immunoglobulin libraries or from animals
transgenic for one or more human immunoglobulin and that do not express
endogenous immunoglobulins, as described infra and, for example in, U.S.
Pat. No. 5,939,598 by Kucherlapati et al.
[0063] The phrase "substantially identical," in the context of two nucleic
acids
or polypeptides (e.g., DNAs encoding a C35 antibody or the amino acid
sequence of the C35 antibody) refers to two or more sequences or

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
subsequences that have at least about 80%, most preferably 90-95% or
higher nucleotide or amino acid residue identity, when compared and
aligned for maximum correspondence, as measured using the following
sequence comparison method and/or by visual inspection. Such
"substantially identical" sequences are typically considered to be
homologous. Preferably, the "substantial identity" exists over a region of
the sequences that is at least about 50 residues in length, more preferably
over a region of at least about 100 residues, and most preferably the
sequences are substantially identical over at least about 150 residues, or
over the full length of the two sequences to be compared. As described
below, any two antibody sequences can only be aligned in one way, by
using the numbering scheme in Kabat. Therefore, for antibodies, percent
identity has a unique and well-defined meaning.
[0064] Amino acids from the variable regions of the mature heavy and light
chains of immunoglobulins are designated Hx and Lx respectively, where
x is a number designating the position of an amino acid according to the
scheme of Kabat, Sequences of Proteins of Immunological Interest
(National Institutes of Health, Bethesda, Md., 1987 and 1991). Kabat lists
many amino acid sequences for antibodies for each subgroup, and lists the
most commonly occurring amino acid for each residue position in that
subgroup to generate a consensus sequence. Kabat uses a method for
assigning a residue number to each amino acid in a listed sequence, and
this method for assigning residue numbers has become standard in the
field. Kabat's scheme is extendible to other antibodies not included in his
compendium by aligning the antibody in question with one of the
consensus sequences in Kabat by reference to conserved amino acids. The
use of the Kabat numbering system readily identifies amino acids at
equivalent positions in different antibodies. For example, an amino acid at
the L50 position of a human antibody occupies the equivalent position to
an amino acid position L50 of a mouse antibody.

CA 02548180 2011-12-28
I.
26
[0065] The basic antibody structural unit is known to
comprise a tetramer.
Each tetramer is composed of two identical pairs of polypeptide chains,
each pair having one "light" (about 25 kDa) and one "heavy" chain (about
50-70 kDa). The amino-terminal portion of each chain includes a variable
region of about 100 to 110 or more amino acids primarily responsible for
antigen recognition. The carboxy-terminal portion of each chain defines a
constant region primarily responsible for effector function. The variable
regions of each light/heavy chain pair form the antibody binding site. Thus,
an intact antibody has two binding sites.
[0066] Light chains are classified as either kappa or lambda.
Heavy chains are
classified as gamma, mu, alpha, delta, or epsilon, and define the antibody's
isotype as IgG, IgM, IgA, IgD and IgE, respectively. Within light and
heavy chains, the variable and constant regions are joined by a "J" region
of about 12 or more amino acids, with the heavy chain also including a "D"
region of about 10 more amino acids. (See generally, Fundamental
Immunology, Paul, W., ed., 3rd ed. Raven Press, N.Y., 1993, SH. 9).
[0067] From N-terminal to C-terminal, both light and heavy
chain variable
regions comprise alternating framework and complementarity determining
regions (CDRs): FR, CDR, FR, CDR, FR, CDR and FR. The assignment
of amino acids to each region is in accordance with the definitions of
Kabat (1987) and (1991), supra, and/or Chothia & Lesk, J. Mol. Biol.
196:901-917 (1987); Chothia et al., Nature 342:878-883 (1989).
[0068] As used herein, the term "framework region" refers to
those portions of
antibody light and heavy chain variable regions that are relatively
conserved (i.e., other than the CDRs) among different immunoglobulins in
a single species, as defined by Kabat, et at., op. cit. As used herein, a
"human framework region" is a framework region that is substantially
identical (about 85% or more) to the framework region of a naturally
occurring human antibody.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
27
[00691 Most preferably for use in humans, the antibodies are human or
humanized antigen-binding antibody fragments of the present invention
and include, but are not limited to, Fab, Fab' and F(ab')2, Fd, single-chain
Fvs (scFv), diabodies, triabodies, tetrabodies, minibodies, domain-deleted
antibodies, single-chain antibodies, disulfide-linked Fvs (sdFv) and
fragments comprising either a VL or VH region. Antigen-binding antibody
fragments, including single-chain antibodies, may comprise the variable
region(s) alone or in combination with the entirety or a portion of the
following: hinge region, CHI, CH2, and CH3 domains. Also included in
the invention are antigen-binding fragments also comprising any
combination of variable region(s) with a hinge region, CH1, CH2, and
CH3 domains.
[0070] Preferred antibodies in the therapeutic methods of the invention are
those containing a deletion of the CH2 domain.
[0071] As used herein, the term "humanized" immunoglobulin or
"humanized" antibody refers to an immunoglobulin comprising a human
framework, at least one CDR from a non-human antibody, and in which
any constant region present is substantially identical to a human
immunoglobulin constant region, i.e., at least about 85-90%, preferably at
least 95% identical. Hence, all parts of a humanized immunoglobulin,
except possibly the CDRs, are substantially identical to corresponding
parts of one or more native human immunoglobulin sequences. For
example, a humanized immunoglobulin would not encompass a chimeric
mouse variable region/human constant region antibody.
[0072] As used herein, the term "chimeric" antibody refers to an antibody
whose heavy and light chains have been constructed, typically by genetic
engineering, from immunoglobulin gene segments belonging to different
species. For example, the variable (V) segments of the genes from a
mouse monoclonal antibody may be joined to human constant (C)
segments, such as gammal and/or gamma4. A typical therapeutic or
diagnostic chimeric antibody is thus a hybrid protein comprising at least

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
28
one V region (e.g., VII or VL) or the entire antigen-binding domain (i.e.,
Vii and VL) from a mouse antibody and at least one C (effector) region
(e.g., CH (CH1, CH2, CH3, or CH4) or CL (CL1, CL2, CL3, or CL4)) or
the entire C domain (i.e., CH and CL) from a human antibody, although
other mammalian species may be used. in some embodiments, especially
for use in the therapeutic methods of the invention, chimeric antibodies
contain no CH2 domain.
[0073] The antibodies of the present invention may be monospecific,
bispecific, trispecific or of greater multispecificity. Multispecific
antibodies may be specific for different epitopes of a polypeptide of the
present invention or may be specific for both a polypeptide of the present
invention as well as for a heterologous epitope, such as a heterologous
polypeptide or solid support material. See, e.g., PCT publications WO
93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., J.
Immunol. 147:60-69 (1991); U.S. Pat. Nos. 4,474,893; 4,714,681;
4,925,648; 5,573,920; 5,601,819; Kostelny et al., J. Immunol. 148:1547-
1553 (1992).
[0074] The description in this section applies to C35 antibodies and to
other
antibodies useful in the method of the invention. Such antibodies may be
conjugated to or complexed with a toxin, as described herein, or may be
unconjugated or tmcomplexed.
C35 Antibodies
[0075] C35 is an antigen differentially expressed in breast cancer and
certain
other tumor types including melanoma, colon carcinoma, ovarian cancer,
and pancreatic cancer. The C35 protein has been shown to be prenylated
and to associate with internal cell membranes but is not detectable on the
surface membrane of viable tumor cells. The inventors have produced a
number of antibodies, including mouse monoclonal antibodies and human
antibodies, that immunospecifically recognize C35 epitopes. The inventors

CA 02548180 2006-06-01
WO 2005/055936 PCT/US2004/040573
29
have also demonstrated that induction of apoptosis in tumor cells by
treatment either with a chemotherapeutic agent or irradiation results in
surface membrane exposure of C35 that permits intact tumor cells to be
recognized by C35-specific antibodies.
[0076] C35 Polynucleotide and amino acid sequences (SEQ ID NOs:1 and 2)
gccgcg atg agc ggg gag ccg ggg cag acg tcc gta gcg ccc cct ccc
Met Ser Gly Glu Pro Gly Gin Thr Ser Val Ala Pro Pro Pro
1 5 10
gag gag gtc gag ccg ggc agt ggg gtc cgc atc gtg gtg gag tac tgt
Glu Glu Val Glu Pro Gly Ser Gly Val Arg Ile Val Val Glu Tyr Cys
15 20 25 30
gaa ccc tgc ggc ttc gag gcg acc tac ctg gag ctg gcc agt gct gtg
Glu Pro Cys Gly Phe Glu Ala Thr Tyr Leu Glu Leu Ala Ser Ala Val
35 40 45
aag gag cag tat ccg ggc atc gag atc gag tcg cgc ctc ggg ggc aca
Lys Glu Gin Tyr Pro Gly Ile Glu Ile Glu Ser Arg Leu Gly Gly Thr
50 55 60
ggt gcc ttt gag ata gag ata aat gga cag ctg gtg ttc tcc aag ctg
Gly Ala Phe Glu Ile Glu Ile Asn Gly Gin Leu Val Phe Ser Lys Leu
65 70 75
gag aat ggg ggc ttt ccc tat gag aaa gat ctc att gag gcc atc cga
Glu Asn Gly Gly Phe Pro Tyr Glu Lys Asp Leu Ile Glu Ala Ile Arg

CA 02548180 2006-06-01
W02005/055936
PCT/US2004/040573
80 85 90
aga gcc agt aat gga gaa acc cta gaa aag atc acc aac agc cgt cct
Arg Ala Ser Asn Gly Glu Thr Leu Glu Lys Ile Thr Asn Ser Arg Pro
95 100 105 110
ccc tgc gtc atc ctg tga
Pro Cys Val Ile Leu
115
[0077] Thus, this
invention also relates to antibodies against C35,
polynucleotides encoding such antibodies, methods of treating C35-
associated cancers using C35 antibodies and polynucleotides, and methods
of detection and diagnosis using C35 antibodies and polynucleotides. Also
provided are vectors and host cells comprising C35 antibody
polynucleotides, and methods of producing C35 antibodies. As described
in more detail herein, the invention also relates to methods using C35
antibodies for cancer treatment, detection, and diagnosis. The description
above under the "Antibodies" section also applies to C35 antibodies
described herein.
[0078] The present invention is further directed to antibody-based
treatment
methods which involve administering C35 antibodies of the invention to a
subject, preferably a mammal, and most preferably a human, for treating
one or more C35 cancers. Therapeutic compounds of the invention
include, but are not limited to, antibodies of the invention (including
fragments, analogs and derivatives thereof as described herein) and nucleic
acids encoding antibodies of the invention (including fragments, analogs
and derivatives thereof as described herein). The antibodies of the
invention can be used to treat, detect or diagnose C35-associated cancers,
including breast, ovarian, colon, pancreatic, and bladder cancers, and

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
31
melanoma. C35 antibodies of the invention may be provided in
pharmaceutically acceptable compositions as known in the art or as
described herein.
[0079] Antibodies of the invention include, but are not limited to,
polyclonal,
monoclonal, multispecific, human, humanized or chimeric antibodies,
single chain antibodies, scFvs, diabodies, triabodies, tetrabodies,
minibodies, domain-deleted antibodies, Fab fragments, F(ab')2 fragments,
fragments produced by a Fab expression library, anti-idiotypic (anti-Id)
antibodies (including, e.g., anti-Id antibodies to antibodies of the
invention), and epitope-binding fragments of any of the above. The term
"antibody," as used herein, refers to immunoglobulin molecules and
immunologically active portions of immunoglobulin molecules, i.e.,
molecules that contain an antigen binding site that immunospecifically
binds an antigen. The immunoglobulin molecules of the invention can be
of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2,
IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.
[0080] Hybridoma cell lines 1F2.4.1 and 1B3.6.1, specific for C35
polypeptides, were prepared using hybridoma technology. (Kohler et al.,
Nature 256:495 (1975); Kohler et al., Eur. J. Immunol. 6:511 (1976);
Kohler et al., Eur. J. Immunol. 6:292 (1976); Hammerling et al., in:
Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, N.Y., pp. 571-
681 (1981)). Briefly, hybridoma cell lines were generated using standard
PEG fusion to the non-secreting myeloma cell line NS-1 (P3/NS1/1-AG4-
1, ATCC #TIB-18) of splenocytes from BALB/c mice immunized with
syngeneic BCA34 fibroblast tumor cells transduced to overexpress C35.
Following PEG fusion to NS-1, the hybridomas were grown in
methylcellulose semi-solid media. Approximately 2 weeks later,
hybridoma colonies were isolated into 96 well plates and individual
supernatants were tested for reactivity with C35 by ELISA, Western blot,
and immunohistochemistry. Positive hybridomas colonies were subcloned
and screened for reactivity twice to ensure clonality. Antibodies were

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
32
isolated from hybridoma supernatants by protein G affinity purification
using standard methods. Antibodies from two hybridoma cell lines, 1F2
and 1B3, specifically bind recombinant C35 protein in ELISA and Western
Blot assays. Antibodies from hybridoma cell line 1F2 also specifically
stain formalin fixed, paraffin embedded C35 positive tumors and cell lines
by immunohistochemistry. In addition, we have developed intracellular
staining flow cytometry assays for quantitative analysis using antibodies
from hybridoma cell line 1F2 conjugated to Alexa-647 flourochrome.
Each of these antibodies is distinct, yet both are specific for C35 protein.
It is possible to immunoprecipitate C35 protein from cell lysates with
either of these antibodies and detect with the other. Competitive binding
ELISA assays suggest that the monoclonal antibodies produced by
hybridoma cell lines 1F2 and 1B3 bind different epitopes of the C35
protein.
[0081] C35 antibodies of the invention include antibodies which
immunospecifically bind a C35 polypeptide, polypeptide fragment, or
variant of SEQ ID NO:2, and/or an epitope, of the present invention (as
determined by immunoassays well known in the art for assaying specific
antibody-antigen binding).
[0082] As used herein the term "isolated" is meant to describe a compound
of
interest (e.g., a C35 antibody) that is in an environment different from that
in which the compound naturally occurs. "Isolated" is meant to include
compounds that are within samples that are substantially enriched for the
compound of interest and/or in which the compound of interest is partially
or substantially purified.
[0083] As used herein, the terms "substantially enriched" and
"substantially
purified" refers to a compound that is removed from its natural
environment and is at least 60% free, preferably 75% free, and most
preferably 90% free from other components with which it is naturally
associated. As used here, an antibody having the "same specificity" as a
reference antibody means the antibody binds the same epitope as the

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
33
reference antibody. The determination of whether an antibody binds the
same epitope as a reference antibody may be performed using the assays
described in the "Assays For Antibody Binding" section below.
[0084] The antibodies derived from mouse hybridoma cell lines discussed
herein are 1F2 and 1B3. Polynucleotides encoding the VL and VH regions
of these antibodies were cloned into TOPO vectors as described in
Example 6, which were deposited with the American Type Culture
Collection ("ATCC") on the date listed in Table 2, and given ATCC
Deposit Numbers listed in Table 2. The ATCC is located at 10801
University Boulevard, Manassas, VA 20110-2209, USA. The ATCC
deposits were made pursuant to the terms of the Budapest Treaty on the
international recognition of the deposit of microorganisms for purposes of
patent procedure.
[0085] Clone 1F2G was deposited at the ATCC on November 11, 2003 and
given ATCC Deposit Number PTA-5639. Clone 1F2K was deposited at
the ATCC on November 11, 2003 and given ATCC Deposit Number PTA-
5640. Clone 1B3G was deposited at the ATCC on November 11, 2003
and given ATCC Deposit Number PTA-5637. Clone 1B3K was deposited
at the ATCC on November 11, 2003 and given ATCC Deposit Number
PTA-5638.
Table 2. Deposited Polynucleotide Clones Encoding Mouse Anti-C35
Variable Regions
Polynucleotide ATCC Accession No. Deposit Date
Clone
1F2G PTA-5639 November 11, 2003
1F2K PTA-5640 November 11, 2003
1B3G PTA-5637 November 11,2003
1B3K PTA-5638 November 11, 2003
[0086] The sequences of the mouse variable region genes and part of the
vector of the deposited clones are set forth below.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
34
Italics = Topo vector sequence (included in deposited clone)
dotted underline ¨ EcoR1 cloning site of Topo vector
Lowercase = 5'untranslated region including generacer primer
ATG Murine signal peptide begin
bold = Frame work regions (FWR)
double underline = CDR1, CDR2, or CDR3
underline = 5' portion of mouse IgG1 or kappa constant region
[0087] 1F2 murine anti-C35 Vgammal gene polynucleotide sequence (from
clone 1F2G)
GAATTTAGCGGCCGCGAATTCGCCCTItgactggagcacg93acactgacataaactaaagaa
9tagaaaacatctctctcattagaggttgatctttgaggaaaacagggtgttgcctaaaggAT
GAAAGTGTTGAGTCTGTTGTACCTGTTGACAGCCATTCCTGGTATCCTGTCTGATGTACAGCT
TCAGGAGTCAGGACCTGGCCTCGTGAAACCTTCTCAGTCTCTGTCTCTCACCTGCTCTGTCAC
TGGCTACTCCATCACCAGTGGTTATTTCTGGAACTGGATCCGG
CDR1
CAGTTTCCAGGGAACAAACTGGAATGGATGGGCTACATAAGCTACGACGGTAGCAATAA
CDR2
CTCCAACCCATCTCTCAAAAATCGAATCTCCTTCACTCGTGACACATCTAAGAACCAGTTTTT
CCTGAAGTTTAATTCTGTGACTACTGACGACTCAGCTGCATATTACTGTACAAGAGGAACTAC
GGGGTTTGCTTACTGGGGCCAAGGGACTCTGGTCACTGTCTCTGCAGCCAA
CDR3
AACGACACCCCCATCTGTCTATCCACTGGCCCCTGGATCTGCTGCCCAAACTAACTCCAAGGG
CGAATTCGTTTAAACCTGCAGGACTAGTCCCTT (SEQ ID NO: 3)
SIGNAL PEPTIDE =18 AA
FR 1 =30 AA
CDR 1 = 6 AA
FR 2 = 14 AA
CDR 2 = 16 AA

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
FR 3 = 32 AA
CDR 3 = 7 AA
FR 4 = 11 AA
[0088] 1F2 VH amino acid sequence (encoded by clone 1F2G)
DVQLQESGPGLVKPSQSLSLTCSVTGYSITSGYFWNWIRQFPGNKLEWMGYISYDGS
NNSNPSLKNRISFTRDTSKNQFFLKFNSVTTDDSAAYYCTRGTTGFAYWGQGTLVTV
SA (SEQ ID NO:4)
[0089] 1F2 murine anti-C35 kappa V gene polynucleotide sequence (from
clone 1F2K)
CGCGAATTCGCCCTTcgactggagcacga9gacact2acatggactgaaagagtagaaaaatt
agetagggaccaaaattcaaagacagaATGGATTTTCAGGTGCAGATTTTCAGCTTCCTGCTA
ATCAGTGCCTCAGTCAGAATGTCCAGAGGACAAATTGTTCTCACCCAGTCTCCAGCAATCATG
TCTGCATCTCCAGGGGAGAAGGTCACCATATCCTGCAGTGCCAGCTCAAGTGTAAGTTACATG
CDR1
AACTGGTACCAGCAGAAGCCAGGATCCTCCCCCAAACCCTGGA
TTTATCACACATCCAACCTGGCTTCTGGAGTCCCTGCTCGCTTCAGTGGCAGTGGGTCT
CDR2
GGGACCTCTTACTCTCTCACAATCAGCAGCATGGAGGCTGAAGATGCTGCCACTTATTACTGC
CAACAGTATCATAGTTACCCACCCACGTTCGGAGGGGGGACCAAGCTGGAAATAA
CDR3
AACGGGCTGATGCTGCACCAACTGTATCCATCTTCCCACCATCCAGTGAGCAAAGGGCGAATT
CGTTT (SEQ ID NO:5)
SIGNAL PEPTIDE = 22 AA
FR1 = 23 AA
CDR 1 = 10 AA
FR 2 = 15 AA
CDR 2 = 7 AA
FR 3 = 32 AA

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
36
CDR 3 = 9 AA
FR 4 = 10 Al
[0090] 1F2-VK amino acid sequence (encoded by clone 1F2K)
QIVLTQSPAIMSASPGEKVTISCSASSSVSYMNWYQQKPGSSPKPWIYHTSNLASGVPARFSG
SGSGTSYSLTISSMEAEDAATYYCQQYHSYPPTFGGGTKLEIK (SEQ ID NO: 6)
[0091] 1B3 murine anti-C35 Vgamma V-gene (encoded by clone 1B3G)
(NC1-A7 V139-D-J1 (VH36-60) M13281)
CGCGAATTCGCCCTTcgactggagcacgaggacactagacatggactgaaggagtagaaaatc
tctctcactggaggctgatttttgaagaaaggggttgtagcctaaaagATGATGGTGTTAAGT
CTTCTGTACCTGTTGACAGCCCTTCCGGGTATCCTGTCAGAGGTGCAGCTTCAGGAGTCAGGA
CCTAGCCTCGTGAAACCTTCTCAGACTCTGTCCCTCACCTGTTCTGTCACTGGCGACTCCATC
ACCAGTGGTTACTGGAACTGGATCCGGAAATTCCCAGGAAATA
CDR1
AACTTGAATACGTGGGGTACATAAGCTACAGTGGTGGCACTTACTACAATCCATCTCTC
CDR2
AAAAGTCGAATCTCCATCACTCGAGACACATCCAAGAACCACTACTACCTGCAGTTGAATTCT
GTGACTACTGAGGACACAGCCACATATTACTGTGCAAGAGGTGCTTACTACGGGGGGGCCTTT
TTTCCTTACTTCGATGTCTGGGGCGCTGGGACCACGGTCACCGTCTCCTCA
CDR3
GCCAAAACGACACCCCCATCTGTCTATCCACTGGCCCCTGGATCTGCTGCCCAAACTAACTCC
AAGGGCGAATTCGTTTAAACCTGC (SEQ ID NO:7)
SIG PEP = 18 AA
FR1 = 30AA
CDR 1 = 5 AA
FR2 = 14 AA
CDR2 = 16 AA
FR 3 = 32 AA

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
37
CDR 3 = 14 AA
FR 4 = 11 AA
[0092] 1B3 VH amino acid sequence (encoded by clone 1B3G)
EVQLQESGPSLVKPSQTLSLTCSVTGDSITSGYWNWIRKFPGNKLEYVGYISYSGGTYYNPSL
KSRISITRDTSKNHYYLQLNSVTTEDTATYYCARGAYYGGAFFPYFDVWGAGTTVTVSS
(SEQ ID NO:8)
[0093] 1B3 murine anti-C35 kappa V-gene (from clone 1B3K)
GAATTCGCCCTTcccetggagcacgaggacactgacatggactgaaggagtagaaaatcagtt
cctgccaggacacagtttagatATGAGGTTCCAGGTTCAGGTTCTGGGGCTCCTTCTGCTCTG
GATATCAGGTGCCCACTGTGATGTCCAGATAACCCAGTCTCCATCTTTTCTTGCTGCATCTCC
TGGAGAAACCATTACTATTAATTGCAGGGCAAGTAAGTACATTAGCA
CDR1
AACATTTAGTCTGGTATCAGGAGAAACCTGGAGAAACTAAAAAGCTTCTTATCTACTCTGGAT
CCACTTTGCAATCTGGACTTCCATCAAGGTTCAGTGGCAGTGGATCTGGTACAGA
CDR2
TTTCACTCTCACCATCAGTAGCCTGGAGCCTGAAGATTTTGCAATGTATTACTGTCAACAGCA
TAATGAATACCCGCTCACGTTCGGTGCTGGGACCAAGCTGGAGCTGAAACGGGCT
CDR3
GATGCTGCACCAACTGTATCCATCTTCCCACCATCCAGTGAGCAAAGGGCGAATTC (SEQ
ID NO:9)
SP = 20AA
FR1=23 aa
CDR1 =11 aa
FR2 = 15 aa
CDR2 = 7 AA
FR3 = 32 aa
CDR 3 = 9 AA
FR 4 = 10 AA

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
38
1B3 VK amino acid sequence (encoded by clone 1B3K)
DVQITQSPSFLAASPGETITINCRASKYISKHLVWYQEKPGETKKLLIYSGSTLQSGLPS
RFSGSGSGTDFTLTISSLEPEDFAMYYCQQHNEYPLTFGAGTKLELK (SEQ ID NO:10)
[0094] The present inventors have also produced two C35 antibodies, MAb
165 and MAb 171, using the method disclosed in US 2002 0123057 Al,
published 5 September 2002. The heavy chain variable regions of MAb
165 and MAb 171 comprise the same CDR3 region as the the 1B3
antibody heavy chain variable region described above. The remainders of
MAbs 165 and 171 are of human origin. The present invention is directed
to antibodies that immunospecifically bind C35 polypeptides, comprising
any one of the VH or VL regions of SEQ ID NO:56, SEQ ID NO:58, or
SEQ ID NO:60, or a combination of either VII region encoded by SEQ ID
NO:56 or SEQ ID NO:60 and the VL region encoded by SEQ II) NO:58,
and preferably the C35-specific antibodies MAb 165 or MAb 171. Both
MAb 165 and MAb 171 comprise the same kappa light chain, UH8 VK
L120.
[0095] The sequences of the heavy and light chain variable regions of MAb
165 and MAb 171 are set forth below.
UNDERLINE = CDR1, CDR2, or CDR3
[0096] MAb 165 VII (141D10 VH H732) nucleotide sequence:
CAGGTGCAGCTGCAGGAGTCGGGCCCAGGACTGGTGAAGCCTCCGGAGACCCTGT
CCCTCACCTGCAATGTCTCTGGTGGCTCTATCGGTAGATACTATTGGAACTGGATC
CDR1
CGACAGTCCCCAGGGAAGGGGCTGGAGTGGATTGGCCATATCCATTACAGTGGGA
GCACCATCTACCATCCCTCCCTCAAGAGTCGAGTCAGCATATCGCTGGACACGTCC
CDR2
AAGAACCAGGTCTCCCTGAAGTTGAGTTCTGTGACCGCTGCGGACACGGCCGTGT
ATTACTGTGCACGAGGTGCTTACTACGGGGGGGCCTTTTTTCCTTACTTCGATGTC
CDR3
TGGGGCCAAGGGACCA CGGTCACCGTCTCCTCA (SEQ ID NO:56)

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
39
[0097] MAb 165 VH (141D10 VH H732) amino acid sequence:
QVQLQESGPGLVKPPETLSLTCNVSGGSIGRYYWNWIRQSPGKGLEWIGHIHYSGSTI
YHPSLKSRVSISLDTSKNQVSLICLSSVTAADTAVYYCARGAYYGGAFFPYFDVWGQG
TTVTVSS (SEQ ID NO:57)
[0098] MAb 171 VH (MSH3 VH H835) nucleotide sequence:
CAGGTGCAGCTGCAGGAGTCGGGAGGAGGCTTAGTTCAGCCTGGGGGGTCCCTGA
GACTCTCTTGTGCAGGCTCTGGATTCACCTTCAGTAGTTACTGGATGCACTGGGTC
CDR1
CGCCAAGCTCCAGGGAAGGGGCTGGTGTGGGTCTCACGTATTGACACTGATGGGA
GTACCACAACCTACGCGGACTCCGTGAAGGGCCGATTCACCATCTCCAGAGACAA
CDR2
CGCCAAGAACACACTGTATCTGCAAATGAACAGCCTGAGAGTCGAGGACACGGCC
GTGTATTACTGTGCACGAGGTGCTTACTACGGGGGGGCCTTTTTTCCTTACTTCGA
CDR3
TGTCTGGGGCCAAGGGACCACGGTCACCGTCTCCTCA (SEQ ID NO:60)
[0100] MAb 171 VH (141D10 VH H732) amino acid sequence:
QVQLQESGGGLVQP GGSLRLS CAGS GFTF S SYWMHWVRQAP GKGINWVSRIDTD GS
TTTYADSVKGRFTISRDNAKNTLYLQMNSLRVEDTAVYYCARGAYYGGAFFPYFDV
WGQGTTVTVSS (SEQ ID NO:61)

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
[0101] UH8 VK L120 nucleotide sequence:
GACATCCAGATGACCCAGTCTCCATCCTCCCTGTCTGCATCTATGGGAGACAGAGT
CACCATCACTTGCCGGGCGAGTCAGGGCATTAGGAATCATTTAGCCTGGTATCAG
CDR1
CAGAAACCAGGGAAAGCTCCTAATCTCCTGATCTCTGCTGCATCCACTTTGCAAT
CDR2
CAGGGGTCCCAACTCGATTCAGTGGCAGTGGATCTGGAACAGATTTCACTCTCAC
CATCAGCAGCCTGCAGCCTGAAGACTCTGCAACTTATTACTGCCAACAGTATAATC
GGTACCCCCTCACTTTCGGCCAA GGGACCAAGCTCGAGATCAAA (SEQ ID NO:58)
CDR3
[0102] UH8 VK L120 amino acid sequence:
DIQMTQ SPSSLSASMGDRVTITCRASOGIRNHLAWYQQIUGICAPNLLISAASTLQSGV
PTRFSGSGSGTDFTLTISSLQPEDSATYYCOOYNRYPLTFGQGTKLEM (SEQ ID
NO:59)
[0103] The present inventors have also produced a human C35 antibody,
MAbc009, using the method disclosed in US 2002 0123057 Al. The present
invention is directed to antibodies that immunospecifically bind C35
polypeptides, comprising the VII and VL regions encoded by the
polynucleotide clones that are listed in Table 3, preferably the fully human
C35-specific antibody MAbc009. Polynucleotides encoding the VL and VH
regions of this antibody were cloned into TOPO vectors as described in
Example 6, which were deposited with the American Type Culture Collection
("ATCC") on the date listed in Table 3, and given ATCC Deposit Numbers
listed in Table 3. The ATCC is located at 10801 University Boulevard,
Manassas, VA 20110-2209, USA. The ATCC deposit was made pursuant to
the terms of the Budapest Treaty on the international recognition of the
deposit
of microorganisms for purposes of patent procedure.
[0104] Clone H0009 was deposited at the ATCC on November 11, 2003 and
given ATCC Deposit Number PTA-5641. Clone L0010 was deposited at the
ATCC on November 11, 2003 and given ATCC Deposit Number PTA-5542.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
41
Table 3. Deposited Polynucleotide Clones Encoding Human Anti-C35
Variable Regions
Polynucleotide Encoded Antibody ATCC Deposit Date
Clone Region Accession No.
H0009 VII of MAbc009 PTA-5641 Nov. 11, 2003
L0010 VL of MAbc009 PTA-5642 Nov. 11, 2003
[01051 The sequences of the human variable region genes and part of the
vector of the deposited clones are set forth below.
DOTTED UNDERLINE = EcoR1 Cloning Site Of Topo Vector
ATG = human signal peptide begin
BOLD = FRAME WORK REGIONS
DOUBLE UNDERLINE = CDR1, CDR2, OR CDR3
UNDERLINE = Human IgG1GS or Kappa Constant Region
[01061 MAbc0009 VH NUCLEOTIDE SEQUENCE (from clone H0009)
GAATTCGCCCTTAATTGCGGCCGCAAACCATGGGATGGAGCTGTATCATCCTC
TTCTTGGTAGCAACAGCTACAGGCGCGCACTCCGAGGTGCAGCTGGTGGAG
TCTGGGGGAGGCGTGGTCCAGCCTGGGAGGTCCCTGAGACTCTCCTGTGC
AGCGTCTGGATTCAACTTCGGTACCTATGCCATGCACTGGGTCCGCCA
CDR1
GGCTCAAGGCAAGGGGCTGGAGTGGGTGGCACTCATATGGTATGATGGAA
CTAAGAAATACTATGCAGACTCCGTGAAGGGCCGATACACCATCTCCAGAG
CDR2
ACAATTCCCAGAACACGCTGTATCTGCAAATGAACACCCTGAGAGCCGAC
GACACGGCTGTGTATTACTGTGCGAAATCAAAACTCCAGGGGCGCGTTA
CDR3
TAGACTACTGGGGCCAGGGAACCCTGGTCACCGTCTCCTCAGCCTCCACCAAGGGCCCATCGG
TCTTCCCCCTGGCACCCTCCTCCAAGAGCACCTCTGGGGGCACAGCGGCCCTGGGCTGCCTGG
TCAAGGACTACTTAAGGGCGAATTC (SEQ ID NO:11)

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
42
[0107] MAbc0009 VII AMINO ACID SEQUENCE (encoded by clone
H0009)
EVQLVESGGGVVQPGRSLRLSCAASGFNFGTYAMHWVRQAQGKGLEWVALIWYDGTKKYYADS
VKGRYTISRDNSQNTLYLQMNTLRADDTAVYYCAKSKL.WRVIDYWGQGTINTVSS (SEQ
ID NO:12)
MAbc0009 VK NUCLEOTIDE SEQUENCE (from clone L0010)
GAATTCGCCCTTAATTGCGGCCGCAAACATGGGATGGAGCTGTATCATCCTCTTCTTGGTAGC
AACAGCTACAGGCGTGCACTCCGACATCCAGATGACCCAGTCTCCAGACtCCCTGGCTGTGTC
TCTGGGCGAGAGGGCCACCATCAACTGCAAGTCCAGCCAGAGTGTTTTATACAGCTCCAACAA
CDR1
TAAGAACTACTTAGCTTGGTACCAGCAGAAACCAGGACAGCCTCCTAAGCTGCTCATTTACTG
GGCATCTACCCGGGAATCCGGGGTCCCTGACCGATTCAGTGGCAGCGGGTCT
CDR2
GGGACAGATTTCACTCTCACCATCAGCAGCCTGCAGGCTGAAGATGTGGCAGTTTATTACTGT
CAGCAATATTATAGTACTCCTCTGTGGACGTTCGGCC
CDR3
AAGGGACCAAGCTCGAGATCAAACGAACTGTGGCTGCACCATCTGTCTTCATCTTCCCGCCAT
CTGATGAGCAGTTGAAATCTGGAACTGCCTCTGTTGTGTGCCTGCTGAAAAGGGCGAATTC
(SEQ ID NO:13)
[0108] MAbc0009 VK AMINO ACID SEQUENCE (encoded by clone
L0010)
IQMTQSPDSLAVSLGERATINCKSSQSVLYSSNNKL\TYLAWYQQKPGQPPKLLIYWASTRESGV
PDRFSGSGSGTDFTLTISSLaAEDVAVYYCQQYYSTPLWTFGQGTKLEIK (SEQ ID
ON:14)
[0109] The mouse C35 antibodies have heavy and light chain variable
regions
designated SEQ ID Nos:3-10. The mouse antibodies 1F2 and 1B3 have
gammal isotype and kappa light chains. The antibodies MAb 165 and MAb
171 that have the same heavy chain variable region CDR3 as 1B3 mouse
antibody have heavy and light chain variable regions designated SEQ ID
NOs:56-60. The antibodies MAb 165 and MAb 171 have kappa light chains.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
43
The human antibody MAbc009 has heavy and light chain variable regions
designated SEQ ID Nos:11-14. The human antibody MAbc009 has gammal
isotype and kappa light chains.
[0110] The present invention encompasses antibodies (including molecules
comprising, or alternatively consisting of, antibody fragments or variants
thereof) that immunospecifically bind to a C35 polypeptide or a fragment,
variant, or fusion protein thereof. A C35 polypeptide includes, but is not
limited to, the C35 polypeptide of SEQ ID NO:2. C35 polypeptides may be
produced through recombinant expression of nucleic acids encoding the
polypeptide of SEQ ID NO:2. (See WO 01/74859 for epitope-containing
fragments of C35.)
[0111] Preferably, analogs of exemplified antibodies differ from
exemplified
antibodies by conservative amino acid substitutions. For purposes of
classifying amino acids substitutions as conservative or nonconservative,
amino acids may be grouped as follows: Group I (hydrophobic sidechains):
met, ala, val, leu, ile; Group II (neutral hydrophilic side chains): cys, ser,
thr;
Group III (acidic side chains): asp, glu; Group IV (basic side chains): asn,
gln,
his, lys, arg; Group V (residues influencing chain orientation): gly, pro; and

Group VI (aromatic side chains): trp, tyr, phe. Conservative substitutions
involve substitutions between amino acids in the same class. Non-conservative
substitutions constitute exchanging a member of one of these classes for a
member of another.
[01121 In one embodiment of the present invention, antibodies that
immunospecifically bind to a C35 polypeptide or a fragment or variant thereof,

comprise a polypeptide having the amino acid sequence of SEQ ID NO:57 or
61, or any one of the VH regions encoded by at least one of the
polynucleotides referred to in Tables 2 or 3 and/or SEQ ID NO:59 or any one
of the VL regions encoded by at least one of the polynucleotides referred to
in
Tables 2 or 3. In further embodiments, antibodies of the present invention
comprise the amino acid sequence of SEQ ID NO:57 or SEQ ID NO:61 and
the amino acid sequence of SEQ ID NO:59. In preferred embodiments,

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
44
antibodies of the present invention comprise the amino acid sequence of a VII
region encoded by clone H0009 and a VL region encoded by clone L0010,
referred to in Table 3. In preferred embodiments, antibodies of the present
invention comprise the amino acid sequence of a VH region encoded by clone
1F2G and a VL region encoded by clone 1F2K, or a VH region encoded by
clone 1B3G and a VL region encoded by clone 1B3K of Table 2. In other
preferred embodiments, antibodies of the present invention comprise the
amino acid sequence of a VH region encoded by clone 110009 of Table 3, the
amino acid sequence of SEQ ID NO:57, or the amino acid sequence of SEQ
ID NO:61 and a VL region encoded by clone 1F2K or 1B3K of Table 2; or a
VH region encoded by clone 1F2G or 1B3G of Table 2, the amino acid
sequence of SEQ ID NO:57, or the amino acid sequence of SEQ ID NO:61
and a VL region encoded by clone L0010 of Table 3; or a VH region encoded
by clone 1F2G or 1B3G of Table 2 or clone 11009 of Table 3 and the amino
acid sequence of SEQ ID NO:59. In other preferred embodiments, antibodies
of the present invention comprise the amino acid sequence of a VH region
encoded by clone 1F2G and a VL region encoded by clone 1B3K of Table 2,
or a VII region encoded by clone 1B3G and a VL region encoded by clone
1F2K of Table 2. Molecules comprising, or alternatively consisting of,
antibody fragments or variants of the VH and/or VL regions encoded by at
least one of the polynucleotides referred to in Tables 2 or 3 that
immunospecifically bind to a C35 polypeptide are also encompassed by the
invention, as are nucleic acid molecules encoding these VH and VL regions,
molecules, fragments and/or variants.
[0113] The present invention also provides antibodies that
immunospecifically
bind to a polypeptide, or polypeptide fragment or variant of a C35
polypeptide,
wherein said antibodies comprise, or alternatively consist of, a polypeptide
having an amino acid sequence of any one, two, three, or more of the VII
CDRs contained in VII regions encoded by one or more polynucleotides of
SEQ ID NOs:56 or 60 or referred to in Tables 2 or 3. In particular, the
invention provides antibodies that immunospecifically bind a C35 polypeptide,

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
comprising, or alternatively consisting of, a polypeptide having the amino
acid
sequence of a VH CDR1 contained in a VII region encoded by one or more
polynucleotides of SEQ ID NOs:56 or 60 or referred to in Tables 2 or 3. In
another embodiment, antibodies that immunospecifically bind a C35
polypeptide, comprise, or alternatively consist of, a polypeptide having the
amino acid sequence of a VII CDR2 contained in a VH region encoded by one
or more polynucleotides of SEQ ID NOs:56 or 60 or referred to in Tables 2 or
3. In a preferred embodiment, antibodies that immunospecifically bind a C35
polypeptide, comprise, or alternatively consist of a polypeptide having the
amino acid sequence of a VII CDR3 contained in a VH region encoded by one
or more polynucleotides of SEQ ID NOs:56 or 60 or referred to in Tables 2 or
3. Molecules comprising, or alternatively consisting of, these antibodies, or
antibody fragments or variants thereof, that immunospecifically bind to C35
polypeptide or a C35 polypeptide fragment or variant thereof are also
encompassed by the invention, as are nucleic acid molecules encoding these
antibodies, molecules, fragments and/or variants.
[0114] The present invention also provides antibodies that
immunospecifically
bind to a polypeptide, or polypeptide fragment or variant of a C35
polypeptide,
wherein said antibodies comprise, or alternatively consist of, a polypeptide
having an amino acid sequence of any one, two, three, or more of the VL
CDRs contained in a VL region encoded by one or more polynucleotides of
SEQ ID NO:58 or referred to in Tables 2 or 3. In particular, the invention
provides antibodies that immunospecifically bind a C35 polypeptide,
comprising, or alternatively consisting of, a polypeptide having the amino
acid
sequence of a VL CDR1 contained in a VL region encoded by one or more
polynucleotides of SEQ ID NO:58 or referred to in Tables 2 or 3. In another
embodiment, antibodies that immunospecifically bind a C35 polypeptide,
comprise, or alternatively consist of, a polypeptide having the amino acid
sequence of a VL CDR2 contained in a VL region encoded by one or more
polynucleotides of SEQ ID NO:58 or referred to in Tables 2 or 3. In a
preferred embodiment, antibodies that immunospecifically bind a C35

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
46
polypeptide, comprise, or alternatively consist of a polypeptide having the
amino acid sequence of a VL CDR3 contained in a VL region encoded by one
or more polynucleotides of SEQ ID NO:58 or referred to in Tables 2 or 3.
Molecules comprising, or alternatively consisting of, these antibodies, or
antibody fragments or variants thereof, that immunospecifically bind to C35
polypeptide or a C35 polypeptide fragment or variant thereof are also
encompassed by the invention, as are nucleic acid molecules encoding these
antibodies, molecules, fragments and/or variants.
[0115] The present invention also provides antibodies (including
molecules
comprising, or alternatively consisting of, antibody fragments or variants)
that
immunospecifically bind to a C35 polypeptide or polypeptide fragment or
variant of a C35 polypeptide, wherein said antibodies comprise, or
alternatively consist of, one, two, three, or more VH CDRs and one, two, three

or more VL CDRs, as contained in a VH region or VL region encoded by one
or more polypeptides of SEQ ID NOs:57, 59, or 61 or referred to in Tables 2
or 3. In
particular, the invention provides for antibodies that
immunospecifically bind to a polypeptide or polypeptide fragment or variant
of a C35 polypeptide, wherein said antibodies comprise, or alternatively
consist of, a VII CDR1 and a VL CDR1, a VII CDR1 and a VL CDR2, a VH
CDR1 and a VL CDR3, a VII CDR2 and a VL CDR1, VII CDR2 and VL
CDR2, a VH CDR2 and a VL CDR3, a VH CDR3 and a VH CDR1, a VH
CDR3 and a VL CDR2, a VH CDR3 and a VL CDR3, or any combination
thereof, of the VH CDRs and VL CDRs contained in a VH region or VL
region encoded by one or more polynucleotides of SEQ ID NOs:56, 58, or 60
or referred to in Tables 2 or 3. The one, two, three, or more VII CDRs and
one, two, three, or more VL CDRs may be from clones H0009 and L0010,
clones H0009 and 1F2K, clones H0009 and 1B3K, clone H009 and SEQ ID
NO:58, clones 1F2G and 1F2K, clones 1F2G and 1B3K, clones 1F2G and
L0010, clone 1F2G and SEQ JD NO:58, clones 1B3G and 1B3K, clones 1B3G
and 1F2K, clones 1B3G and L0010, clone 1B3G and SEQ ID NO:58, SEQ ID
NO:56 and SEQ ID NO:58, SEQ ID NO:56 and clone L0010, SEQ ID NO:56

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
47
and clone 1F2K, SEQ ID NO:56 and clone 1B3K, SEQ ID NO:60 and SEQ ID
NO:58, SEQ ID NO:60 and clone L0010, SEQ ID NO:60 and clone 1F2K, or
SEQ ID NO:60 and clone 1B3K. Molecules comprising, or alternatively
consisting of, fragments or variants of these antibodies, that
immunospecifically bind to C35 polypeptide are also encompassed by the
invention, as are nucleic acid molecules encoding these antibodies, molecules,

fragments or variants.
[0116] Most preferably the antibodies are human, chimeric (e.g.,
human.mouse chimeric), or humanized antibodies or antigen-binding antibody
fragments of the present invention, including, but not limited to, Fab, Fab'
and
F(ab')2, Fd, single-chain Fvs (scFv), diabodies, triabodies, tetrabodies,
minibodies, single-chain antibodies, disulfide-linked Fvs (sdFv), and
intrabodies, and fragments comprising either a VL or VH region. Antigen-
binding antibody fragments, including single-chain antibodies, may comprise
the variable region(s) alone or in combination with the entirety or a portion
of
the following: hinge region, CHI, CH2, and CH3 domains. Also included in
the invention are antigen-binding fragments also comprising any combination
of variable region(s) with a hinge region, CHL CH2, and CH3 domains.
Preferred C35 antibodies in the therapeutic methods of the invention are those

containing a deletion of the CH2 domain.
[0117] Antibodies of the present invention may be described or specified
in
terms of the epitope(s) or portion(s) of a polypeptide of the present
invention
which they recognize or specifically bind. The epitope(s) or polypeptide
portion(s) may be specified as described herein, e.g., by N-terminal and C-
terminal positions, or by size in contiguous amino acid residues. Antibodies
which specifically bind any epitope or polypeptide of the present invention
may also be excluded. Therefore, the present invention includes antibodies
that
specifically bind polypeptides of the present invention, and allows for the
exclusion of the same.
[0118] Antibodies of the present invention may also be described or
specified
in terms of their binding affinity to a polypeptide of the invention.
Preferred

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
48
binding affinities include those with a dissociation constant or Kd less than
5
X 10(-7) M, 10(-7) M, 5 X 10(-8) M, 10(-8) M, 5 X 10(-9) M, 10(-9) M, 5 X
10(40) M, 10(-10) M, 5 X 10(-11) M, 10(-11) M, 5 X 10(42) M, 10(42) M,
X 10(-13) M, 10(-13) M, 5 X 10(-14) M, 10(44) M, 5 X 10(45) M, or 10(-
15) M.
[0119] Antibodies of the invention have an affinity for C35 the same as or
similar to the affinity of the antibodies 1F2, 1B3, MAb 165, MAb 171, or
MAbc009. Preferably, the antibodies of the invention have an affinity for C35
that is higher than the affinity of the antibodies 1F2, 1B3, MAb 165, MAb
171, or MAbc009.
[0120] The invention also provides antibodies that competitively inhibit
binding of an antibody to a C35 epitope as determined by any method known
in the art for determining competitive binding, for example, the immunoassays
and antibody binding assays described herein. In preferred embodiments, the
antibody competitively inhibits binding to the epitope by at least 95%, at
least
90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or
at
least 50%.
[0121] Antibodies of the present invention may also be described or
specified
in terms of their cross-reactivity. Antibodies that do not bind any other
analog,
ortholog, or homolog of a polypeptide of the present invention are included.
Antibodies that bind polypeptides with at least 95%, at least 90%, at least
85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at
least 55%, and at least 50% identity (as calculated using methods known in the

art and described herein) to a polypeptide of the present invention are also
included in the present invention. In specific embodiments, antibodies of the
present invention cross-react with murine, rat and/or rabbit homologs of
human proteins and the corresponding epitopes thereof Antibodies that do not
bind polypeptides with less than 95%, less than 90%, less than 85%, less than
80%, less than 75%, less than 70%, less than 65%, less than 60%, less than
55%, and less than 50% identity (as calculated using methods known in the art
and described herein) to a polypeptide of the present invention are also

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
49
included in the present invention. In a specific embodiment, the above-
described cross-reactivity is with respect to any single specific antigenic or

immunogenic polypeptide, or combination(s) of 2, 3, 4, 5, or more of the
specific antigenic and/or immunogenic polypeptides disclosed herein. Further
included in the present invention are antibodies which bind polypeptides
encoded by polynucleotides which hybridize to a polynucleotide of the present
invention under stringent hybridization conditions (as described herein).
[0122] Antibodies of the present invention may be described or specified
in
terms of the epitope(s) or portion(s) of a polypeptide of the present
invention
which they recognize or specifically bind. The epitope(s) or polypeptide
portion(s) may be specified as described herein, e.g., by N-terminal and C-
terminal positions, by size in contiguous amino acid residues, or listed in
the
Tables and Figures. Antibodies which specifically bind any epitope or
polypeptide of the present invention may also be excluded. Therefore, the
present invention includes antibodies that specifically bind polypeptides of
the
present invention, and allows for the exclusion of the same. Excluded from
the invention are antibodies against C35 disclosed in US 2002/0052308 and/or
WO 01/74859, and/or antibodies that specifically bind an epitope disclosed
therein.
[0123] In a specific embodiment, antibodies of the present invention bind
to
an epitope contained within the fragment represented by residues 105 to 115 of

the native C35 sequence. In another embodiment, antibodies of the present
invention bind to an epitope contained within the fragment represesnted by
residues 53-104 of the native C35 sequence.
[0124] Antibodies of the present invention may also be described or
specified
in terms of their cross-reactivity, or lack thereof. Antibodies that do not
bind
any other analog, ortholog, or homolog of a polypeptide of the present
invention are included. Antibodies that bind polypeptides with at least 95%,
at
least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least
65%,
at least 60%, at least 55%, and at least 50% identity (as calculated using
methods known in the art and described herein) to a polypeptide of the present

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
invention are also included in the present invention. In specific embodiments,

antibodies of the present invention cross-react with murine, monkey, rat
and/or
rabbit homologs of human proteins and the corresponding epitopes thereof.
Antibodies that do not bind polypeptides with less than 95%, less than 90%,
less than 85%, less than 80%, less than 75%, less than 70%, less than 65%,
less than 60%, less than 55%, and less than 50% identity (as calculated using
methods known in the art and described herein) to a polypeptide of the present
invention are also included in the present invention. In a
specific
embodiment, the above-described cross-reactivity is with respect to any single

specific antigenic or immunogenic polypeptide, or combination(s) of 2, 3, 4,
5,
or more of the specific antigenic and/or immunogenic polypeptides disclosed
herein. Further included in the present invention are antibodies which bind
polypeptides encoded by polynucleotides which hybridize to a polynucleotide
of the present invention under stringent hybridization conditions (as
described
herein).
[0125] In preferred embodiments, the antibodies of the present
invention
(including molecules comprising, or alternatively consisting of, antibody
fragments or variants thereof), immunospecifically bind to C35 polypeptide
and do not cross-react with any other antigens.
[0126] In a
preferred embodiment, antibodies of the invention preferentially
bind C35 polypeptide (SEQ ID NO:2), or fragments and variants thereof
relative to their ability to bind other antigens.
[0127] By way of non-limiting example, an antibody may be considered to
bind a first antigen preferentially if it binds said first antigen with a
dissociation constant (KD) that is less than the antibody's KD for the second
antigen. In another non-limiting embodiment, an antibody may be considered
to bind a first antigen preferentially if it binds said first antigen with an
affinity
that is at least one order of magnitude less than the antibody's KD for the
second antigen. In another non-limiting embodiment, an antibody may be
considered to bind a first antigen preferentially if it binds said first
antigen

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
51
with an affinity that is at least two orders of magnitude less than the
antibody's
KD for the second antigen.
[0128] In another non-limiting embodiment, an antibody may be considered
to
bind a first antigen preferentially if it binds said first antigen with an off
rate
(k(off)) that is less than the antibody's k(off) for the second antigen. In
another non-limiting embodiment, an antibody may be considered to bind a
first antigen preferentially if it binds said first antigen with an affinity
that is at
least one order of magnitude less than the antibody's k(off) for the second
antigen. In another non-limiting embodiment, an antibody may be considered
to bind a first antigen preferentially if it binds said first antigen with an
affinity
that is at least two orders of magnitude less than the antibody's k(off) for
the
second antigen.
[0129] In specific embodiments, antibodies of the invention bind C35
polypeptides or fragments or variants thereof with an off rate (k(off)) of
less
than or equal to 5 X 10(-2) sec-1, 10(-2) sec-1, 5 X 10(-3) sec-1 or 10(-3)
sec-1.
More preferably, antibodies of the invention bind C35 polypeptides or
fragments or variants thereof with an off rate (k(off)) less than or equal to
5 X
10(-4) sec-1, 10(-4) sec-1, 5 X 10(-5) sec-1, or 10(-5) sec-1 5 X 10(-6) sec-
1,
10(-6) sec-1, 5 X 10(-7) sec-1 or 10(-7) sec-1.
[0130] In other embodiments, antibodies of the invention bind C35
polypeptides or fragments or variants thereof with an on rate (k(on)) of
greater
than or equal to 10(3) M-1 sec-1, 5 X 10(3) M-1 sec-1, 10(4) M-1 sec-1 or 5 X
10(4) M-1 sec-1. More preferably, antibodies of the invention bind C35
polypeptides or fragments or variants thereof with an on rate (k(on)) greater
than or equal to 10(5) M-1 sec-1, 5 X 10(5) M-1 sec-1, 10(6) M-1 sec-1, or 5
X 10(6)M-1 sec-1 or 10(7) M-1 sec-1.
[0131] The present invention also provides antibodies that comprise, or
alternatively consist of, variants (including derivatives) of the antibody
molecules (e.g., the VH regions and/or VL regions) described herein, which
antibodies immunospecifically bind to a C35 polypeptide or fragment or
variant thereof. Standard techniques known to those of skill in the art can be

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
52
used to introduce mutations in the nucleotide sequence encoding a molecule of
the invention, including, for example, site-directed mutagenesis and PCR-
mediated mutagenesis which result in amino acid substitutions. Preferably, the

variants (including derivatives) encode less than 50 amino acid substitutions,

less than 40 amino acid subsitutions, less than 30 amino acid substitutions,
less
than 25 amino acid substitutions, less than 20 amino acid substitutions, less
than 15 amino acid substitutions, less than 10 amino acid substitutions, less
than 5 amino acid substitutions, less than 4 amino acid substitutions, less
than
3 amino acid substitutions, or less than 2 amino acid substitutions relative
to
the reference VH region, VHCDR1, VHCDR2, VHCDR3, VL region,
VLCDR1, VLCDR2, or VLCDR3. A "conservative amino acid substitution"
is one in which the amino acid residue is replaced with an amino acid residue
having a side chain with a similar charge. Families of amino acid residues
having side chains with similar charges have been defined in the art. These
families include amino acids with basic side chains (e.g., lysine, arginine,
histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged

polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine,
tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine,
isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched
side
chains ( e.g., threonine, valine, isoleucin.e) and aromatic side chains (e.g.,

tyrosine, phenylalanine, tryptophan, histidine). Alternatively, mutations can
be
introduced randomly along all or part of the coding sequence, such as by
saturation mutagenesis, and the resultant mutants can be screened for
biological activity to identify mutants that retain activity (e.g., the
ability to
bind a C35 polypeptide).
[0132] For example, it is possible to introduce mutations only in
framework
regions or only in CDR regions of an antibody molecule. Introduced
mutations may be silent or neutral missense mutations, i.e., have no, or
little,
effect on an antibody's ability to bind antigen. These types of mutations may
be useful to optimize codon usage, or improve a hybridoma's antibody
production. Alternatively, non-neutral missense mutations may alter an

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
53
antibody's ability to bind antigen. The location of most silent and neutral
missense mutations is likely to be in the framework regions, while the
location
of most non-neutral missense mutations is likely to be in CDR, though this is
not an absolute requirement. One of skill in the art would be able to design
and test mutant molecules with desired properties such as no alteration in
antigen binding activity or alteration in binding activity (e.g., improvements
in
antigen binding activity or change in antibody specificity). Following
mutagenesis, the encoded protein may routinely be expressed and the
functional and/or biological activity of the encoded protein, (e.g., ability
to
immunospecifically bind a C35 polypeptide) can be determined using
techniques described herein or by routinely modifying techniques known in the
art.
[0133] In a specific embodiment, an antibody of the invention (including
a
molecule comprising, or alternatively consisting of, an antibody fragment or
variant thereof), that immunospecifically binds C35 polypeptides or fragments
or variants thereof, comprises, or alternatively consists of, an amino acid
sequence encoded by a nucleotide sequence that hybridizes to a nucleotide
sequence that is complementary to that encoding one of the VH or VL regions
encoded by one or more of the nucleic acids of SEQ ID NOs:56, 58, or 60 or
referred to in Tables 2 or 3 under stringent conditions, e.g., hybridization
to
filter-bound DNA in 6X sodium chloride/sodium citrate (SSC) at about 45 C
followed by one or more washes in 0.2xSSC/0.1% SDS at about 50-65 C,
under highly stringent conditions, e.g., hybridization to filter-bound nucleic

acid in 6xSSC at about 45 C followed by one or more washes in
0.1xSSC/0.2% SDS at about 68 C, or under other stringent hybridization
conditions which are known to those of skill in the art (see, for example,
Ausubel, F.M. et al., eds. , 1989, Current Protocols in Molecular Biology,
Vol.
1, Green Publishing Associates, Inc. and John Wiley & Sons, Inc., New York
at pages 6.3.1-6.3.6 and 2.10.3). Nucleic acid molecules encoding these
antibodies are also encompassed by the invention.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
54
[0134] It is well known within the art that polypeptides, or fragments or
variants thereof, with similar amino acid sequences often have similar
structure and many of the same biological activities. Thus, in one
embodiment, an antibody (including a molecule comprising, or alternatively
consisting of, an antibody fragment or variant thereof), that
immunospecifically binds to a C35 polypeptide or fragments or variants of a
C35 polypeptide, comprises, or alternatively consists of, a VH region having
an amino acid sequence that is at least 35%, at least 40%, at least 45%, at
least
50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at
least 80%, at least 85%, at least 90%, at least 95%, or at least 99%
identical, to
the amino acid sequence of a VH region encoded by a nucleic acid of SEQ ID
NOs:56 or 60 or referred to in Tables 2 or 3.
[0135] In another embodiment, an antibody (including a molecule
comprising,
or alternatively consisting of, an antibody fragment or variant thereof), that

immunospecifically binds to a C35 polypeptide or fragments or variants of a
C35 polypeptide, comprises, or alternatively consists of, a VL region having
an amino acid sequence that is at least 35%, at least 40%, at least 45%, at
least
50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at
least 80%, at least 85%, at least 90%, at least 95%, or at least 99%
identical, to
the amino acid sequence of a VL region encoded by a nucleic acid of SEQ ID
NO:58 or referred to in Tables 2 or 3.
[0136] The invention also encompasses antibodies (including molecules
comprising, or alternatively consisting of, antibody fragments or variants
thereof) that have one or more of the same biological characteristics as one
or
more of the antibodies described herein. By "biological characteristics" is
meant, the in vitro or in vivo activities or properties of the antibodies,
such as,
for example, the ability to bind to C35 polypeptide (e.g., C35 polypeptide
expressed on a cell surface during apoptosis); the ability to substantially
inhibit
or abolish C35 polypeptide mediated biological activity; the ability to kill
C35-
associated cancer cells (e.g., treat or diagnose C35-associated cancer), or
detect C35; the ability to inhibit or abolish C35 polypeptide mediated

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
biological activity. Optionally, the antibodies of the invention will bind to
the
same epitope as at least one of the antibodies specifically referred to
herein.
Such epitope binding can be routinely determined using assays known in the
art.
[0137] The rules described below for producing humanized antibodies
derived
from mouse VH and VL regions encoded by the nucleic acids referred to in
Table 2 may also be used to produce antibody variants comprising the human
VH and/or VL regions encoded by SEQ ID NOs: 56, 58, or 60 or by the
nucleic acids referred to in Table 3.
[0138] Humanized immunoglobulins and human antibody variants of the
invention have variable framework regions substantially from a human
immunoglobulin (termed an acceptor immunoglobulin), and CDRs
substantially from the mouse C35 VH and VL regions encoded by the clones
in Table 2 or from the human C35 VH and VL regions encoded by the clones
in Table 3 (referred to as the donor immunoglobulin). The constant region(s),
if present, are also substantially from a human immunoglobulin. The
humanized antibodies and human antibody variants exhibit a specific binding
affinity for C35 of at least 10(2), 10(3), 10(4), 10(5), 10(6), 10(7), 10(8),
10(9), or 10(10) M(-1). Usually the upper limit of binding affinity of the
humanized antibodies and human antibody variants for human C35 is within a
factor of 3, 4, 5 or 10 of that of the mouse antibodies 1F2 or 1B3 or the
human
antibody MAbc009, or of antibodies MAb 165 or MAb 171. Often the lower
limit of binding affinity is also within a factor of 3, 4, 5 or 10 of that of
the
mouse antibodies in 1F2 or 1B3 or human antibody MAbc009, or of
antibodies MAb 165 or MAb 171. Preferred humanized immunoglobulins and
human antibody variants compete with the mouse antibodies 1F2 or 1B3 or
human antibody MAbc009, or antibodies MAb 165 or MAb 171 for binding to
C35 and prevent C35 from binding to the respective mouse or human
antibody.
[0139] The heavy and light chain variable regions of possible human
acceptor
antibodies are described by Kabat, Sequences of Proteins of Immunological

CA 02548180 2011-12-28
56
Interest (National Institutes of Health, Bethesda, Md., 1987 and 1991). The
human acceptor antibody is chosen such that its variable regions exhibit a
high
degree of sequence identity with those of the mouse C35 antibody. The heavy
and light chain variable framework regions can be derived from the same or
different human antibody sequences. The human antibody sequences can be
the sequences of naturally occurring human antibodies or can be consensus
sequences of several human antibodies.
[0140] The design of humanized immunoglobulins can be carried out as
follows. When an amino acid falls under the following category, the
framework amino acid of a human immunoglobulin to be used (acceptor
immunoglobulin) is replaced by a framework amino acid from a CDR-
providing non-human immunoglobulin (donor immunoglobulin):
[0141] (a) the amino acid in the human framework region of the acceptor
immunoglobulin is unusual for human immunoglobulins at that position,
whereas the corresponding amino acid in the donor immunoglobulin is typical
for human immunoglobulins in that position;
[0142] (b) the position of the amino acid is immediately adjacent to
one of the
CDRs; or
[0143] (c) the amino acid is capable of interacting with the CDRs (see,
Queen
et al. WO 92/11018., and Co et al., Proc. Natl. Acad. Sci. USA 88, 2869
(1991), respectively). For a
detailed description of the production of humani7ed immunoglobulins see,
Queen et al. and Co et al.
[0144] Usually the CDR regions in humanized antibodies and human
antibody
variants are substantially identical, and more usually, identical to the
corresponding CDR regions in the mouse or human antibody from which they
were derived. Although not usually desirable, it is sometimes possible to make

one or more conservative amino acid substitutions of CDR residues without
appreciably affecting the binding affinity of the resulting humanized
immunoglobulin or human antibody variant. Occasionally, substitutions of
CDR regions can enhance binding affinity.

CA 02548180 2011-12-28
57
[0145] Other than for the specific amino acid substitutions discussed
above,
the framework regions of humanized immunoglobulins and human antibody
variants are usually substantially identical, and more usually, identical to
the
framework regions of the human antibodies from which they were derived
(acceptor immunoglobulin). Of course, many of the amino acids in the
framework region make little or no direct contribution to the specificity or
affinity of an antibody. Thus, many individual conservative substitutions of
framework residues can be tolerated without appreciable change of the
specificity or affinity of the resulting humanized immunoglobulin or human
antibody variants.
[0146] For example, analogs of HuZAF show substantial amino acid sequence
identity with HuZAF. Heavy and light chain variable regions of analogs are
encoded by nucleic acid sequences that hybridize with the nucleic acids
encoding the heavy or light chain variable regions of HuZAF, or degenerate
forms thereof, under stringent conditions. Phage-display technology offers
powerful techniques for selecting such analogs while retaining binding
affinity
and specificity (see, e.g., Dower et al., WO 91/17271; McCafferty et al., WO
92/01047; and Huse, WO 92/06204.
[0147] The VH and VL genes in the nucleic acid clones in Tables 2 or 3 or
SEQ ID NO:s 56, 58, or 60 can be employed to select fully human antibodies
specific for C35 according to the method taught by US 2002 0123057A1, "In
vitro methods of producing and identifying immunoglobulin molecules in
eukaryotic cells," published 5 September 2002. Briefly, the mouse (or human)
VII linked to a human CH is employed to select fully human immunoglobulin
light chains from a library of such light chains that when paired with the
mouse (or human) VH confers specificity for C35. The selected fully human
immunoglobulin light chains are then employed to select fully human
immunoglobulin heavy chains from a library of such heavy chains that when
paired with the fully human light chain confer specificity for C35. Similarly,
= the mouse (or human) VL linked to a human CL may be employed to select

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
58
fully human immunoglobulin heavy chains from a library of such heavy chains
that when paired with the mouse (or human) VL confers specificity for C35.
The selected fully human immunoglobulin heavy chains are then employed to
select fully human immunoglobulin light chains from a library of such light
chains that when paired with the fully human heavy chain confer specificity
for
C35. Frequently, the fully human antibody selected in this fashion has epitope

specificity that is identical or closely related to that of the original mouse
(or
human) C35-specific antibody.
[0148] The method of US 2002 0123057 Al may also be used with a library
of heavy or light chains of which all members have one or more non-human
(e.g., mouse) CDRs. In one example, each member of the library comprises a
CDR3 region derived from an isolated murine monoclonal antibody specific
for C35, e.g., 1F2 or 1B3.
[0149] All fully human antibodies or antibodies having one or more non-
human (e.g., mouse) CDRs (including molecules comprising, or alternatively
consisting of, antibody fragments or variants thereof) selected through use of

the method of US 2002 0123057 Al starting with immunoglobulin heavy or
light chain variable regions encoded by the nucleic acids of SEQ ED NO:s 56,
58, or 60 or referred to in Tables 2 or 3 are encompassed in the present
invention.
[0150] The variable segments of humanized antibodies or human antibody
variants produced as described supra are typically linked to at least a
portion of
an immunoglobulin constant region (Fc), typically that of a human
immunoglobulin. Human constant region DNA sequences can be isolated in
accordance with well-known procedures from a variety of human cells, such as
immortalized B-cells (see Kabat et al., supra, and WO 87/02671). The
antibody may contain both light chain and heavy chain constant regions. The
heavy chain constant region may include CH1, hinge, CH2, CH3, and,
sometimes, CH4 regions. For therapeutic purposes, the CH2 domain may be
deleted or omitted.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
59
[0151] The humanized antibody or human antibody variants include
antibodies having all types of constant regions, including IgM, IgG, IgD, IgA
and IgE, and any isotype, including IgGl, IgG2, IgG3 and IgG4. When it is
desired that the humanized antibody or human antibody variants exhibit
cytotoxic activity, the constant domain is usually a complement-fixing
constant domain and the class is typically IgG1 . When such cytotoxic activity

is not desirable, the constant domain can be of the IgG2 class. The humanized
antibody or human antibody variants may comprise sequences from more than
one class or isotype.
[0152] Chimeric antibodies are also encompassed in the present invention.
Such antibodies may comprise the VH region and/or VL region encoded by the
nucleic acids of SEQ ID NOs:56, 58, or 60 or in Tables 2 or 3 fused to the CH
region and/or CL region of a another species, such as human or mouse or
horse, etc. In preferred embodiments, a chimeric antibody comprises the VII
and/or VL region encoded by the nucleic acids of Table 2 fused to human C
regions. The human CH2 domain may be deleted when antibodies are used in
therapeutic purposes. Chimeric antibodies encompass antibody fragments, as
described above.
[0153] The variable segments of chimeric antibodies are typically linked
to at
least a portion of an immunoglobulin constant region (Fc), typically that of a

human immunoglobulin. Human constant region DNA sequences can be
isolated in accordance with well-known procedures from a variety of human
cells, such as immortalized B-cells (see Kabat et al., supra, and WO
87/02671). The antibody may contain both light chain and heavy chain
constant regions. The heavy chain constant region may include CH1, hinge,
CH2, CH3, and, sometimes, CH4 regions. For therapeutic purposes, the CH2
domain may be deleted or omitted.
[0154] The variable segments of chimeric antibodies produced as described
supra are typically linked to at least a portion of an immunoglobulin constant

region (Fc), typically that of a human immunoglobulin. Human constant region
DNA sequences can be isolated in accordance with well-known procedures

CA 02548180 2011-12-28
=
from a variety of human cells, such as immortalized B-cells (see Kabat et al.,

supra, and WO 87/02671). The antibody may contain both light chain and
heavy chain constant regions. The heavy chain constant region may include
CH1, hinge, CH2, CH3, and, sometimes, CH4 regions. For therapeutic
purposes, the CH2 domain may be deleted or omitted.
[0155] Chimeric antibodies include antibodies having all types of
constant
regions, including IgM, IgG, IgD, IgA and IgE, and any isotype, including
IgGl, IgG2, IgG3 and IgG4. When it is desired that the chimeric antibody
exhibit cytotoxic activity, the constant domain is usually a complement-fixing

constant domain and the class is typically IgGl. When such cytotoxic activity
is not desirable, the constant domain can be of the IgG2 class. The chimeric
antibody may comprise sequences from more than one class or isotype.
[0156] A variety of methods are available for producing such.
immunoglobulins. Because of the degeneracy of the genetic code, a variety of
nucleic acid sequences encode each immunoglobulin amino acid sequence.
The desired nucleic acid sequences can be produced by de novo solid-phase
DNA synthesis or by PCR mutagenesis of an earlier prepared variant of the
desired polynucleotide. All nucleic acids encoding the antibodies described in

this application are expressly included in the invention.
[0157] Once expressed, the whole antibodies, their dimers, individual
light
and heavy chains, or other immunoglobulin forms of the present invention can
be purified according to standard procedures in the art, including ammonium
sulfate precipitation, affinity columns, column chromatography, gel
electrophoresis and the like (see, generally, Scopes, R., Protein
Purification,
Springer-Verlag, N.Y. (1982)).
Substantially pure immunoglobulins of at least about 90 to 95% homogeneity
are preferred, and 98 to 99% or more homogeneity most preferred, for
pharmaceutical uses. Once purified, partially or to homogeneity as desired,
the
polypeptides may then be used therapeutically (including extracorporeally), in

developing and performing assay procedures, immunofluorescent stainings,
and the like. (See, generally, Immunological Methods, Vols. I and II,
Lefkovits

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
61
and Pemis, eds., Academic Press, New York, N.Y. (1979 and 1981), or detect
C35 or diagnose a C35-associated cancer.
[0158] The present invention also provides for fusion proteins
comprising, or
alternatively consisting of, an antibody (including molecules comprising, or
alternatively consisting of, antibody fragments or variants thereof), that
immunospecifically binds to C35 polypeptide, and a heterologous polypeptide.
Preferably, the heterologous polypeptide to which the antibody is fused is
useful for function or is useful to target the C35 polypeptide expressing
cells,
including but not limited to breast, ovarian, bladder, colon, and pancreatic
cancer cells, and melanoma cells. In an alternative preferred embodiment, the
heterologous polypeptide to which the antibody is fused is useful for T cell,
macrophage, and/or monocyte cell function or is useful to target the antibody
to a T cell, macrophage, or monocyte. In one embodiment, a fusion protein of
the invention comprises, or alternatively consists of, a polypeptide having
the
amino acid sequence of any one or more of the VH regions of an antibody of
the invention or the amino acid sequence of any one or more of the VL regions
of an antibody of the invention or fragments or variants thereof, and a
heterologous polypeptide sequence. In another embodiment, a fusion protein
of the present invention comprises, or alternatively consists of, a
polypeptide
having the amino acid sequence of any one, two, three, or more of the VH
CDRs of an antibody of the invention, or the amino acid sequence of any one,
two, three, or more of the VL CDRs of an antibody of the invention, or
fragments or variants thereof, and a heterologous polypeptide sequence. In a
preferred embodiment, the fusion protein comprises, or alternatively consists
of, a polypeptide having the amino acid sequence of a VH CDR3 of an
antibody of the invention, or fragment or variant thereof, and a heterologous
polypeptide sequence, which fusion protein immunospecifically binds to C35
polypeptide. In another embodiment, a fusion protein comprises, or
alternatively consists of a polypeptide having the amino acid sequence of at
least one VH region of an antibody of the invention and the amino acid
sequence of at least one VL region of an antibody of the invention or

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
62
fragments or variants thereof, and a heterologous polypeptide sequence.
Preferably, the VH and VL regions of the fusion protein correspond to a single

antibody (or scFv or Fab fragment) of the invention. In yet another
embodiment, a fusion protein of the invention comprises, or alternatively
consists of a polypeptide having the amino acid sequence of any one, two,
three or more of the VH CDRs of an antibody of the invention and the amino
acid sequence of any one, two, three or more of the VL CDRs of an antibody
of the invention, or fragments or variants thereof, and a heterologous
polypeptide sequence. Preferably, two, three, four, five, six, or more of the
VHCDR(s) or VLCDR(s) correspond to single antibody (or scFv or Fab
fragment) of the invention. Nucleic acid molecules encoding these fusion
proteins are also encompassed by the invention.
[0159] As discussed in more detail below, the antibodies of the present
invention may be used either alone or in combination with other compositions.
The antibodies may further be recombinantly fused to a heterologous
polypeptide at the N- or C-terminus or chemically conjugated (including
covalent and non-covalent conjugations) to polypeptides or other
compositions. For example, antibodies of the present invention may be
recombinantly fused or conjugated to molecules useful as labels in detection
assays and effector molecules such as heterologous polypeptides, drugs,
radionuclides, or toxins. See, e.g., PCT publications WO 92/08495; WO
91/14438; WO 89/12624; U.S. Patent No. 5,314,995; and EP 396,387.
[0160] By way of another non-limiting example, antibodies of the invention
may be administered to individuals as a form of passive immunization.
Alternatively, antibodies of the present invention may be used for epitope
mapping to identify the epitope(s) bound by the antibody. Epitopes identified
in this way may, in turn, for example, be used as vaccine candidates, i.e., to

immunize an individual to elicit antibodies against the naturally occurring
forms of C35 for therapeutic methods.
[0161] Antibodies of the present invention may act as agonists or
antagonists
of the C35 polypeptides.

CA 02548180 2011-12-28
63
[0162] Antibodies of the present invention may be used, for example, but
not
limited to, to purify, detect, and target the polypeptides of the present
invention, including both in vitro and in vivo diagnostic and therapeutic
methods. For example, the antibodies have use in immunoassays for
qualitatively and quantitatively measuring levels of the polypeptides of the
present invention in biological samples. See, e.g., Harlow et al., Antibodies:
A
Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988).
[0163] The antibodies of the invention include derivatives that are
modified,
i.e, by the covalent attachment of any type of molecule to the antibody such
that covalent attachment does not prevent the antibody from generating an
anti-idiotypic response or binding C35. For example, but not by way of
limitation, the antibody derivatives include antibodies that have been
modified, e.g., by glycosylation, acetylation, pegylation, phosphylation,
phosphorylation, amidation, derivatization by known protecting/blocking
groups, proteolytic cleavage, linkage to a cellular ligand or other protein,
etc.
Any of numerous chemical modifications may be carried out by known
techniques, including, but not limited to specific chemical cleavage,
acetylation, formylation, metabolic synthesis of tunicamycin, etc.
Additionally,
the derivative may contain one or more non-classical amino acids.
[0164] Antibodies of the invention can be composed of amino acids joined to
each other by peptide bonds or modified peptide bonds, i.e., peptide
isosteres,
and may contain amino acids other than the 20 gene-encoded amino acids.
The C35 antibodies may be modified by natural processes, such as
posttranslational processing, or by chemical modification techniques which are

well known in the art. Such modifications are well described in basic texts
and in more detailed monographs, as. well as in a voluminous research
literature. Modifications can occur anywhere in the C35 antibody, including
the peptide backbone, the amino acid side-chains and the amino or carboxyl
termini. It will be appreciated that the same type of modification may be
present in the same or varying degrees at several sites in a given C35
antibody.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
64
Also, a given C35 antibody may contain many types of modifications. C35
antibodies may be branched, for example, as a result of ubiquitination, and
they may be cyclic, with or without branching. Cyclic, branched, and
branched cyclic C35 antibodies may result from posttranslation natural
processes or may be made by synthetic methods. Modifications include
acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of
Ravin, covalent attachment of a heme moiety, covalent attachment of a
nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid
derivative, covalent attachment of phosphotidylinositol, cross-linking,
cyclization, disulfide bond formation, demethylation, formation of covalent
cross-links, formation of cysteine, formation of pyroglutamate, formylation,
gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation,
iodination, methylation, myristoylation, oxidation, pegylation, proteolytic
processing, phosphorylation, prenylation, racemization, selenoylation,
sulfation, transfer-RNA mediated addition of amino acids to proteins such as
arginylation, and ubiquitination. (See, for
instance, PROTEINS -
STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E.
Creighton, W. H. Freeman
and Company, New York (1993);
POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS,
B. C. Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et
al., Meth Enzymol 182:626-646 (1990); Rattan et al., Ann NY Acad Sci
663:48-62 (1992).)
[0165] A further embodiment of the invention relates to a polypeptide
which
comprises the amino acid sequence of a C35 antibody sequence having an
amino acid sequence which contains at least one amino acid substitution, but
not more than 50 amino acid substitutions, even more preferably, not more
than 40 amino acid substitutions, still more preferably, not more than 30
amino
acid substitutions, and still even more preferably, not more than 20 amino
acid
substitutions. Of course, in order of ever-increasing preference, it is highly

preferable for a polypeptide to have an amino acid sequence which comprises
a C35 antibody sequence, which contains at least one, but not more than 10, 9,

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
8, 7, 6, 5, 4, 3, 2 or 1 amino acid substitutions. In specific embodiments,
the
number of additions, substitutions, and/or deletions in the C35 antibody
sequence is 1-5, 5-10, 5-25, 5-50, 10-50 or 50-150. For substitutions,
conservative amino acid substitutions are preferable. The substitutions may be

within the framework regions or the CDRs or both.
[0166] The description in this section applies to C35 antibodies and to
other
antibodies useful in the method of the invention. Such antibodies may be
conjugated to or complexed with a toxin, as described herein, or may be
unconjugated or uncomplexed.
Pol3mucleotides Encoding C35 Antibodies
[0167] The present invention also provides for nucleic acid molecules
encoding a C35 antibody of the invention (including molecules comprising, or
alternatively consisting of, antibody fragments or variants thereof).
[0168] The invention also encompasses polynucleotides that hybridize
under
stringent or alternatively, under lower stringency hybridization conditions,
e.g.,
as defined supra, to polynucleotides that encode an antibody, preferably, that

specifically binds to a C35 polypeptide.
[0169] In a specific embodiment, a nucleic acid molecule of the invention
encodes an antibody (including molecules comprising, or alternatively
consisting of, antibody fragments or variants thereof), comprising, or
alternatively consisting of, a VII region having an amino acid sequence of any

one of the VH regions encoded by a nucleic acid of the invention and a VL
region having an amino acid sequence of any one of the VL regions encoded
by a nucleic acid of the invention. In another embodiment, a nucleic acid
molecule of the invention encodes an antibody (including molecules
comprising, or alternatively consisting of, antibody fragments or variants
thereof), comprising, or alternatively consisting of, a VII region having an
amino acid sequence of any one of the VH regions encoded by a nucleic acid

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
66
of the invention or a VL region having an amino acid sequence of any one of
the VL regions encoded by a nucleic acid of the invention.
[0170] The polynucleotides may be obtained, and the nucleotide sequence of
the polynucleotides determined, by any method known in the art. For example,
if the nucleotide sequence of the antibody is known, a polynucleotide encoding

the antibody may be assembled from chemically synthesized oligonucleotides
(e.g., as described in Kutmeier et al., BioTechniques 17:242 (1994)), which,
briefly, involves the synthesis of overlapping oligonucleotides containing
portions of the sequence encoding the antibody, annealing and ligating of
those
oligonucleotides, and then amplification of the ligated oligonucleotides by
PCR.
[0171] Alternatively, a polynucleotide encoding an antibody may be
generated
from nucleic acid from a suitable source. If a clone containing a nucleic acid

encoding a particular antibody is not available, but the sequence of the
antibody molecule is known, a nucleic acid encoding the immunoglobulin may
be chemically synthesized or obtained from a suitable source (e.g., an
antibody
cDNA library, or a cDNA library generated from, or nucleic acid, preferably
poly A+RNA, isolated from, any tissue or cells expressing the antibody, such
as hybridoma cells selected to express an antibody of the invention) by PCR
amplification using synthetic primers hybridizable to the 3' and 5' ends of
the
sequence or by cloning using an oligonucleotide probe specific for the
particular gene sequence to identify, e.g., a cDNA clone from a cDNA library
that encodes the antibody. Amplified nucleic acids generated by PCR may then
be cloned into replicable cloning vectors using any method well known in the
art.
[0172] Once the nucleotide sequence and corresponding amino acid sequence
of the antibody is determined, the nucleotide sequence of the antibody may be
manipulated using methods well known in the art for the manipulation of
nucleotide sequences, e.g., recombinant DNA techniques, site directed
mutagenesis, PCR, etc. (see, for example, the techniques described in
Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d Ed., Cold

CA 02548180 2011-12-28
67
Spring Harbor Laboratory, Cold Spring Harbor, N.Y. and Ausubel et al., eds.,
1998, Current Protocols in Molecular Biology, John Wiley & Sons, NY),
= to generate
antibodies having a different amino acid sequence, for example to create
amino acid substitutions, deletions, and/or insertions.
[0173] In a specific embodiment, the amino acid sequence of the heavy
and/or
light chain variable domains may be inspected to identify the sequences of the

complementarity determining regions (CDRs) by methods that are well know
in the art, e.g., by comparison to known amino acid sequences of other heavy
and light chain variable regions to determine the regions of sequence
hypervariability. Using routine recombinant DNA techniques, one or more of
the CDRs may be inserted within framework regions, e.g., into human
framework regions to humanize a non-human antibody, as described supra.
The framework regions may be naturally occurring or consensus framework
regions, and preferably human framework regions (see, e.g., Chothia et al., J.

Mol. Biol. 278: 457-479 (1998) for a listing of human framework regions).
Preferably, the polynucleotide generated by the combination of the framework
regions and CDRs encodes an antibody that specifically binds C35. Preferably,
as discussed supra, one or more amino acid substitutions may be made within
the framework regions, and, preferably, the amino acid substitutions improve
binding of the antibody to its antigen. Additionally, such methods may be used

to make amino acid substitutions or deletions of one or more variable region
cysteine residues participating in an intrachain disulfide bond to generate
antibody molecules lacking one or more intrachain disulfide bonds. Other
alterations to the polynucleotide are encompassed by the present invention and

within the skill of the art.
[0174] In addition, techniques developed for the production of "chimeric
antibodies" (Morrison et al., Proc. Natl. Acad. Sci. 81:851-855 (1984);
Neuberger et al., Nature 312:604-608 (1984); Takeda et al., Nature 314:452-
454 (1985)) by splicing genes from a mouse antibody molecule of appropriate
antigen specificity together with genes from a human antibody molecule of

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
68
appropriate biological activity can be used. As described supra, a chimeric
antibody is a molecule in which different portions are derived from different
animal species, such as those having a variable region derived from a murine
mAb and a human immunoglobulin constant region, e.g., humanized
antibodies.
[0175] Alternatively, techniques described for the production of single
chain
antibodies (U.S. Pat. No. 4,946,778; Bird, Science 242:423-42 (1988); Huston
et aL, Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988); and Ward et al.,
Nature 334:544-54 (1989)) can be adapted to produce single chain antibodies.
Single chain antibodies are formed by linking the heavy and light chain
fragments of the Fv region via an amino acid bridge, resulting in a single
chain
polyp eptide. Techniques for the assembly of functional Fv fragments in E.
coli
may also be used (Skerra et al., Science 242:1038-1041 (1988)).
[0176] The present invention also provides for nucleic acid molecules
encoding an antibody of the invention (including molecules comprising, or
alternatively consisting of, antibody fragments or variants thereof). In a
specific embodiment, a nucleic acid molecule of the invention encodes an
antibody (including molecules comprising, or alternatively consisting of,
antibody fragments or variants thereof), comprising, or alternatively
consisting
of, a VH region having an amino acid sequence of any one of the VH regions
encoded by a nucleic acid of SEQ ID NO:s 56 or 60 or referred to in Table 2 or

3 and a VL region having an amino acid sequence of any one of the VL
regions encoded by a nucleic acid of SEQ ID NO:58 or referred to in Table 2
or 3. In another embodiment, a nucleic acid molecule of the invention encodes
an antibody (including molecules comprising, or alternatively consisting of,
antibody fragments or variants thereof), comprising, or alternatively
consisting
of, a VH region having an amino acid sequence of any one of the VH regions
encoded by a nucleic acid of SEQ ID NOs:56 or 60 or referred to in Table 2 or
3 or a VL region having an amino acid sequence of any one of the VL regions
encoded by a nucleic acid of SEQ ID NO:58 or referred to in Table 2 or 3.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
69
[01771 The present invention also provides antibodies that comprise, or
alternatively consist of, variants (including derivatives) of the antibody
molecules (e.g., the VH regions and/or VL regions) described herein, which
antibodies immunospecifically bind to a C35 polypeptide or fragment or
variant thereof. Standard techniques known to those of skill in the art can be

used to introduce mutations in the nucleotide sequence encoding a molecule of
the invention, including, for example, site-directed mutagenesis and PCR-
mediated mutagenesis which result in amino acid substitutions. Preferably, the

variants (including derivatives) encode less than 50 amino acid substitutions,

less than 40 amino acid subsitutions, less than 30 amino acid substitutions,
less
than 25 amino acid substitutions, less than 20 amino acid substitutions, less
than 15 amino acid substitutions, less than 10 amino acid substitutions, less
than 5 amino acid substitutions, less than 4 amino acid substitutions, less
than
3 amino acid substitutions, or less than 2 amino acid substitutions relative
to
the reference VH region, VHCDR1, VHCDR2, VHCDR3, and/or VL region,
VLCDR1, VLCDR2, or VLCDR3. A "conservative amino acid substitution"
is one in which the amino acid residue is replaced with an amino acid residue
having a side chain with a similar charge. Families of amino acid residues
having side chains with similar charges have been defined in the art. These
families include amino acids with basic side chains (e.g., lysine, arginine,
histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged

polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine,
tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine,
isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched
side
chains ( e.g., threonine, valine, isoleucine) and aromatic side chains (e.g.,
tyrosine, phenylalanine, tryptophan, histidine). Alternatively, mutations can
be
introduced randomly along all or part of the coding sequence, such as by
saturation mutagenesis, and the resultant mutants can be screened for
biological activity to identify mutants that retain activity (e.g., the
ability to
bind a C35 polypeptide).

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
[0178] For example, it is possible to introduce mutations only in
framework
regions or only in CDR regions, or both regions, of an antibody molecule.
Introduced mutations may be silent or neutral missense mutations, i.e., have
no, or little, effect on an antibody's ability to bind antigen. These types of

mutations may be useful to optimize codon usage, or improve a hybridoma's
antibody production. Alternatively, non-neutral missense mutations may alter
an antibody's ability to bind antigen. The location of most silent and neutral

missense mutations is likely to be in the framework regions, while the
location
of most non-neutral missense mutations is likely to be in CDR, though this is
not an absolute requirement. One of skill in the art would be able to design
and test mutant molecules with desired properties such as no alteration in
antigen binding activity or alteration in binding activity (e.g., improvements
in
antigen binding activity or change in antibody specificity). Following
mutagenesis, the encoded protein may routinely be expressed and the
functional and/or biological activity of the encoded protein, (e.g., ability
to
immunospecifically bind a C35 polypeptide) can be determined using
techniques described herein or by routinely modifying techniques known in the
art.
[01.79] In a specific embodiment, an antibody of the invention (including a
molecule comprising, or alternatively consisting of, an antibody fragment or
variant thereof), that immunospecifically binds C35 polypeptides or fragments
or variants thereof, comprises, or alternatively consists of, an amino acid
sequence encoded by a nucleotide sequence that hybridizes to a nucleotide
sequence that is complementary to that encoding one of the VH or VL regions
encoded by a nucleic acid of SEQ ID NO:s 56, 58, or 60 or referred to in Table

2 or 3 under stringent conditions, e.g., hybridization to filter-bound DNA in
6X sodium chloride/sodium citrate (SSC) at about 45 C followed by one or
more washes in 0.2xSSC/0.1% SDS at about 50-65 C, under highly stringent
conditions, e.g., hybridization to filter-bound nucleic acid in 6xSSC at about

45 C followed by one or more washes in 0.1xSSC/0.2% SDS at about 68 C,
or under other stringent hybridization conditions which are known to those of

CA 02548180 2011-12-28
71
skill in the art (see, for example, Ausubel, F.M. et al., eds. , 1989, Current

Protocols in Molecular Biology, Vol. I, Green Publishing Associates, Inc. and
John Wiley & Sons, Inc., New York at pages 6.3.1-6.3.6 and 2.10.3). Nucleic
acid molecules encoding these antibodies are also encompassed by the
invention.
[0180] Note that variations in the above conditions may be accomplished
through the inclusion and/or substitution of alternate blocking reagents used
to
suppress background in hybridization experiments. Typical blocking reagents
TM
include Denhardt's reagent, BLOTTO, heparin, denatured salmon sperm DNA,
and commercially available proprietary formulations. The inclusion of
specific blocking reagents may require modification of the hybridization
conditions described above, due to problems with compatibility.
[0181] Of course, a polynucleotide which hybridizes only to polyA+
sequences, or to a complementary stretch of T (or U) residues, would not be
included in the definition of "polynucleotide," since such a polynucleotide
would hybridize to any nucleic acid molecule containing a poly (A) stretch or
the complement thereof (e.g., practically any double-stranded cDNA clone
generated using oligo dT as a primer).
[0182] The C35 antibody polynucleotide can be composed of any
polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA
or DNA or modified RNA or DNA. For example, C35 antibody
polynucleotides can be composed of single- and double-stranded DNA, DNA
that is a mixture of single- and double-stranded regions, single- and double-
stranded RNA, and RNA that is mixture of single- and double-stranded
regions, hybrid molecules comprising DNA and RNA that may be single-
stranded or, more typically, double-stranded or a mixture of single- and
double-stranded regions. In addition, the C35 antibody polynucleotides can be
composed of triple-stranded regions comprising RNA or DNA or both RNA
and DNA. C35 antibody polynucleotides may also contain one or more
modified bases or DNA or RNA backbones modified for stability or for other
reasons. "Modified" bases include, for example, tritylated bases and unusual

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
72
bases such as inosine. A variety of modifications can be made to DNA and
RNA; thus, "polynucleotide" embraces chemically, enzymatically, or
metabolically modified forms.
[0183] Antibodies of the invention (including antibody fragments or
variants)
can be produced by any method known in the art. For example, it will be
appreciated that antibodies in accordance with the present invention can be
expressed in cell lines other than hybridoma cell lines. Sequences encoding
the
cDNAs or genomic clones for the particular antibodies can be used for
transformation of a suitable mammalian or nonmammalian host cells or to
generate phage display libraries, for example. Additionally, polypeptide
antibodies of the invention may be chemically synthesized or produced
through the use of recombinant expression systems.
[0184] One way to produce the antibodies of the invention would be to
clone
the VH and/or VL regions encoded by any one or more of the nucleic acids of
SEQ ID NOs: 56, 58, or 60 or referred to in Table 2 or 3. In order to isolate
the VH and VL regions from the hybridoma cell lines or nucleic acids, PCR
primers including VH or VL nucleotide sequences, may be used to amplify the
VH and VL sequences contained in the vectors containing the nucleic acids of
SEQ ID NOs: 56, 58, or 60 or Table 2 or 3. The PCR products may then be
cloned using vectors, for example, which have a PCR product cloning site
consisting of a 5' and 3' single T nucleotide overhang, that is complementary
to the overhanging single adenine nucleotide added onto the 5' and 3' end of
PCR products by many DNA polymerases used for PCR reactions. The VH
and VL regions can then be sequenced using conventional methods known in
the art.
[0185] The cloned VH and VL genes may be placed into one or more suitable
expression vectors. By way of non-limiting example, PCR primers including
VH or VL nucleotide sequences, a restriction site, and a flanking sequence to
protect the restriction site may be used to amplify the VH or VL sequences.
Utilizing cloning techniques known to those of skill in the art, the PCR
amplified VH regions may be cloned into vectors expressing the appropriate

CA 02548180 2011-12-28
73
immunoglobulin constant region, e.g., the human IgG1 or IgG4 constant
region for VII regions, and the human kappa or lambda constant regions for
kappa and lambda VL regions, respectively. Preferably, the vectors for
expressing the VH or VL regions comprise a promoter suitable to direct
expression of the heavy and light chains in the chosen expression system, a
secretion signal, a cloning site for the immunoglobulin variable domain,
immunoglobulin constant domains, and a selection marker such as neomycin.
The VII and VL regions may also be cloned into a single vector expressing the
necessary constant regions. The heavy chain conversion vectors and light
chain conversion vectors are then co-transfected into cell lines to generate
stable or transient cell lines that express full-length antibodies, e.g., IgG,
using
techniques known to those of skill in the art (See, for example, Guo et al.,
J.
Clin. Endocrinol. Metab. 82:925-31(1997), and Ames et al., J. Imm.unol.
Methods 184:177-86 (1995) ).
[0186] The invention further provides polynucleotides comprising a
nucleotide
sequence encoding an antibody of the invention and fragments thereof. The
invention also encompasses polynucleotides that hybridize under stringent or
lower stringency hybridization conditions, e.g., as defined supra, to
polynucleotides that encode an antibody, preferably, that specifically binds
to a
polypeptide of the invention, preferably, an antibody that binds to a
polypeptide having the amino acid sequence of SEQ ID NO:2 or a polypeptide
encoded by the deposited clone.
[01871 For some uses, such as for in vitro affinity maturation of an
antibody of
the invention, it may be useful to express the VH and VL regions of the heavy
and light chains of one or more antibodies of the invention as single chain
antibodies or Fab fragments in a phage display library. For example, the
cDNAs encoding the VH and VL regions of one or more antibodies of the
invention may be expressed in all possible combinations using a phage display
library, allowing for the selection of VH/VL combinations that bind a C35
polypeptides with preferred binding characteristics such as improved affinity

CA 02548180 2011-12-28
74
or improved off rates. Additionally, VH and VL segments, the CDR regions
of the VII and VL regions of one or more antibodies of the invention, in
particular, may be mutated in vitro. Expression of VII and VL regions with
"mutant" CDRs in a phage display library allows for the selection of VH/VL
combinations that bind a C35 polypeptide receptor polypeptides with preferred
binding characteristics such as improved affinity or improved off rates.
[01881 In phage display methods, functional antibody domains are
displayed
on the surface of phage particles which carry the polynucleofide sequences
encoding them. In particular, DNA sequences encoding VH and VL regions
are amplified from animal cDNA libraries (e.g., human or murine cDNA
libraries of lymphoid tissues) or synthetic cDNA libraries. The DNA encoding
the VH and VL regions are joined together by an scFv linker by PCR and
cloned into a phagemid vector (e.g., p CANTAB 6 or pComb 3 HSS). The
vector is electroporated in E. coli and the E. coli is infected with helper
phage.
Phage used in these methods are typically filamentous phage including fd and
M13 and the VII and VL regions are usually recombinantly fused to either the
phage gene III or gene VIII. Phage expressing an antigen binding domain that
binds to an antigen of interest (i.e., a C35 polypeptide or a fragment
thereof)
can be selected or identified with antigen, e.g., using labeled antigen or
antigen
bound or captured to a solid surface or bead. Examples of phage display
methods that can be used to make the antibodies of the present invention
include, but are not limited to, those disclosed in Brinkman et al., J.
Immunol.
Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177-186
(1995); Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994); Persic et
al.,
Gene 187 9-18 (1997); Burton et al., Advances in Immunology 57:191-
280(1994); PCT application No. PCT/GB91/01 134; PCT publications WO
90/02809; WO 91/10737; WO 92/01047; WO 92/18719; WO 93/1 1236; WO
95/15982; WO 95/20401; W097/13844; and U.S. Patent Nos. 5,698,426;
5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047; 5,571,698;
5,427,908; 5,516,717; 5,780,225; 5,658,727; 5,735,743 and 5,969,108.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
[0189] Alternatively, a preferred method for increasing the affinity of
antibodies of the invention is disclosed in US 2002 0123057 Al.
[0190] In addition, techniques developed for the production of "chimeric
antibodies" (Morrison et al., Proc. Natl. Acad. Sci. 81:851-855 (1984);
Neuberger et al., Nature 312:604-608 (1984); Takeda et at., Nature 314:452-
454 (1985)) by splicing genes from a mouse antibody molecule of appropriate
antigen specificity together with genes from a human antibody molecule of
appropriate biological activity can be used. As described supra, a chimeric
antibody is a molecule in which different portions are derived from different
animal species, such as those having a variable region derived from a murine
mAb and a human immunoglobulin constant region, e.g., humanized
antibodies.
[0191] Alternatively, techniques described for the production of single
chain
antibodies (U.S. Patent No. 4,946,778; Bird, Science 242:423- 42 (1988);
Huston et aL, Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988); and Ward et
al., Nature 334:544-54 (1989)) can be adapted to produce single chain
antibodies. Single chain antibodies are formed by linking the heavy and light
chain fragments of the Fv region via an amino acid bridge, resulting in a
single chain polypeptide. Techniques for the assembly of functional Fv
fragments in E. coli may also be used (Skerra et al., Science 242:1038- 1041
(1988)).
Assays For Antibody Binding
[0192] The antibodies of the invention may be assayed for immunospecific
binding by any method known in the art. The inununoassays which can be
used include but are not limited to competitive and non-competitive assay
systems using techniques such as western blots, radioimmunoassays, ELISA
(enzyme linked immunosorbent assay), "sandwich" immunoassays,
imrnunoprecipitation assays, precipitin reactions, gel diffusion precipitin
reactions, immunodiffusion assays, agglutination assays, complement-fixation

CA 02548180 2011-12-28
76
assays, immtmoradiometric assays, fluorescent immunoassays, protein A
immunoassays, to name but a few. Such assays are routine and well known in
the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular
Biology, Vol. 1, John Wiley & Sons, Inc., New York).
Exemplary immunoassays are described
briefly below (but are not intended by way of limitation).
[0193] lmmunoprecipitation protocols generally comprise lysing a population
of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1%
sodium deoxycholate, 0.1% SDS, 0.15 M NaCI, 0.01 M sodium phosphate at
pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease
inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the
antibody of interest to the cell lysate, incubating for a period of time
(e.g., 1-4
hours) at 4° C., adding protein A and/or protein G sepharose beads to
the cell lysate, incubating for about an hour or more at 4° C., washing

the beads in lysis buffer and resuspending the beads in SDS/sample buffer.
The ability of the antibody of interest to immunoprecipitate a particular
antigen can be assessed by, e.g., western blot analysis. One of skill in the
art
would be knowledgeable as to the parameters that can be modified to increase
the binding of the antibody to an antigen and decrease the background (e.g.,
pre-clearing the cell lysate with sepharose beads). For further discussion
regarding immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994,
Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New
York at 10.16.1.
101941 Western blot analysis generally comprises preparing protein samples,
electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%-20%
SDS-PAGE depending on the molecular weight of the antigen), transferring
the protein sample from the polyacrylamide gel to a membrane such as
nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution
(e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing
buffer (e.g., PBS-Tween 20), blocking the membrane with primary antibody
(the antibody of interest) diluted in blocking buffer, washing the membrane in

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
77
washing buffer, blocking the membrane with a secondary antibody (which
recognizes the primary antibody, e.g., an anti-human antibody) conjugated to
an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase)
or radioactive molecule (e.g., 32P or 1251) diluted in blocking buffer,
washing
the membrane in wash buffer, and detecting the presence of the antigen. One
of skill in the art would be knowledgeable as to the parameters that can be
modified to increase the signal detected and to reduce the background noise.
For further discussion regarding western blot protocols see, e.g., Ausubel et
al,
eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley &
Sons, Inc., New York at 10.8.1.
[0195] ELISAs comprise preparing antigen, coating the well of a 96 well
microtiter plate with the antigen, adding the antibody of interest conjugated
to
a detectable compound such as an enzymatic substrate (e.g., horseradish
peroxidase or alkaline phosphatase) to the well and incubating for a period of

time, and detecting the presence of the antigen. In ELISAs the antibody of
interest does not have to be conjugated to a detectable compound; instead, a
second antibody (which recognizes the antibody of interest) conjugated to a
detectable compound may be added to the well. Further, instead of coating the
well with the antigen, the antibody may be coated to the well. In this case, a

second antibody conjugated to a detectable compound may be added following
the addition of the antigen of interest to the coated well. One of skill in
the art
would be knowledgeable as to the parameters that can be modified to increase
the signal detected as well as other variations of ELISAs known in the art.
For
further discussion regarding ELISAs see, e.g., Ausubel et al, eds, 1994,
Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New
York at 11.2.1.
[0196] The binding affinity of an antibody to an antigen and the off-rate
of an
antibody-antigen interaction can be determined by competitive binding assays.
One example of a competitive binding assay is a radioimmunoassay
comprising the incubation of labeled antigen (e.g., 3H or 1251) with the
antibody of interest in the presence of increasing amounts of unlabeled

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
78
antigen, and the detection of the antibody bound to the labeled antigen. The
affinity of the antibody of interest for a particular antigen and the binding
off-
rates can be determined from the data by scatchard plot analysis. Competition
with a second antibody can also be determined using radioimmunoassays. In
this case, the antigen is incubated with antibody of interest conjugated to a
labeled compound (e.g., 3H or 1251) in the presence of increasing amounts of
an unlabeled second antibody.
[01971 Characterization of the affinity and kinetics of monoclonal
antibody
(Mab) interactions with a specific antigen can be used to probe the structure
and function of either the antibody or the antigen. Kinetic measurements are
also important in the selection of appropriate reagents for immunoassay.
[01981 There are a variety of methods available for measuring the affinity
of
an antibody-antigen interaction, but relatively few for determining rate
constants. Most of the methods rely on either labeling antibody or antigen,
which inevitably complicates routine measurements and introduces
uncertainties in the measured quantities.
[0199] Surface plasmon reasonance (SPR) as performed on BIAcore offers a
number of advantages over conventional methods of measuring the affinity of
antibody-antigen interactions: (i) no requirement to label either antibody or
antigen; (ii) antibodies do not need to be purified in advance, cell culture
supernatant can be used directly; (iii) real-time measurements, allowing rapid

semi-quantitative comparison of different monoclonal antibody interactions,
are enabled and are sufficient for many evaluation purposes; (iv) biospecific
surface can be regenerated so that a series of different monoclonal antibodies

can easily be compared under identical conditions; (v) analytical procedures
are fully automated, and extensive series of measurements can be performed
without user intervention. BIAapplications Handbook, version AB (reprinted
1998), BIACORE code No. BR-1001-86; BlAtechnology Handbook, version
AB (reprinted 1998), BIACORE code No. BR-1001-84.
[0200] SPR based binding studies require that one member of a binding pair
be immobilized on a sensor surface. The binding partner immobilized is

CA 02548180 2011-12-28
79
referred to as the ligand. The binding partner in solution is referred to as
the
analyte. In some cases, the ligand is attached indirectly to the surface
through
binding to another immobilized molecule, which is referred as the capturing
molecule. SPR response reflects a change in mass concentration at the detector

surface as analytes bind or dissociate.
TM
[0201] Based on SPR, real-time BlAcore measurements monitor interactions
directly as they happen. The technique is well suited to determination of
kinetic parameters. Comparative affinity ranking is extremely simple to
perform, and both kinetic and affmity constants can be derived from the
sensorgram data.
[0202] When analyte is injected in a discrete pulse across a ligand
surface, the
resulting sensorgram can be divided into three essential phases: (i)
Association
of analyte with ligand during sample injection; (ii) Equilibrium or steady
state
during sample injection, where the rate of analyte binding is balanced by
dissociation from the complex; (iii) Dissociation of analyte from the surface
during buffer flow.
[0203] The association and dissociation phases provide information on the
kinetics of analyte-ligand interaction (ka and kd, the rates of complex
formation
and dissociation, kd/ka = KD). The equilibrium phase provides information on
the affinity of the analyte-ligand interaction (Ka
[0204] BIAevaluatiorisoftware provides comprehensive facilities for curve
fitting using both numerical integration and global fitting algorithms. With
suitable analysis of the data, separate rate and affinity constants for
interaction
can be obtained from simple BIAcorrinvestigations. The range of affinities
measurable by this technique is very broad ranging from mM to pM.
[0205] Epitope specificity is an important characteristic of a monoclonal
antibody. Epitope mapping with BlAcoreT,m in contrast to conventional
techniques using radioimmunoassay, ELISA or other surface adsorption
methods, does not require labeling or purified antibodies, and allows multi-
site
specificity tests using a sequence of several monoclonal antibodies.
Additionally, large numbers of analyses can be processed automatically.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
[0206] Pair-wise binding experiments test the ability of two MAbs to bind
simultaneously to the same antigen. MAbs directed against separate epitopes
will bind independently, whereas MAbs directed against closely related
epitopes will interfere with each other's binding. These binding experiments
with BlAcore are straightforward to carry out.
[0207] For example, one can use a capture molecule to bind the first Mab,
followed by addition of antigen and second MAb sequentially. The
sensorgrams will reveal: 1. how much of the antigen binds to first Mab, 2. to
what extent the second MAb binds to the surface-attached antigen, 3. if the
second MAb does not bind, whether reversing the order of the pair-wise test
alters the results.
[0208] Peptide inhibition is another technique used for epitope mapping.
This
method can complement pair-wise antibody binding studies, and can relate
functional epitopes to structural features when the primary sequence of the
antigen is known. Peptides or antigen fragments are tested for inhibition of
binding of different MAbs to immobilized antigen. Peptides which interfere
with binding of a given MAb are assumed to be structurally related to the
epitope defined by that MAb.
Methods Of Producing Antibodies
[0209] The antibodies of the invention can be produced by any method known
in the art for the synthesis of antibodies, in particular, by chemical
synthesis or
preferably, by recombinant expression techniques.
[021.0] Recombinant expression of an antibody of the invention, or
fragment,
derivative or analog thereof, (e.g., a heavy or light chain of an antibody of
the
invention or a single chain antibody of the invention), may require
construction of an expression vector containing a polynucleotide that encodes
the antibody. Once a polynucleotide encoding an antibody molecule or a heavy
or light chain of an antibody, or portion thereof (preferably containing the
heavy or light chain variable domain), of the invention has been obtained, the

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
81
vector for the production of the antibody molecule may be produced by
recombinant DNA technology using techniques well known in the art. Thus,
methods for preparing a protein by expressing a polynucleotide containing an
antibody encoding nucleotide sequence are described herein. Methods which
are well known to those skilled in the art can be used to construct expression

vectors containing antibody coding sequences and appropriate transcriptional
and translational control signals. These methods include, for example, in
vitro
recombinant DNA techniques, synthetic techniques, and in vivo genetic
recombination. The invention, thus, provides replicable vectors comprising a
nucleotide sequence encoding an antibody molecule of the invention, or a
heavy or light chain thereof, or a heavy or light chain variable domain,
operably linked to a promoter. Such vectors may include the nucleotide
sequence encoding the constant region of the antibody molecule (see, e.g.,
PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Pat.
No. 5,122,464) and the variable domain of the antibody may be cloned into
such a vector for expression of the entire heavy or light chain.
[0211] The expression vector is transferred to a host cell by
conventional
techniques and the transfected cells are then cultured by conventional
techniques to produce an antibody of the invention. Thus, the invention
includes host cells containing a polynucleotide encoding an antibody of the
invention, or a heavy or light chain thereof, or a single chain antibody of
the
invention, operably linked to a heterologous promoter. In preferred
embodiments for the expression of double-chained antibodies, vectors
encoding both the heavy and light chains may be co-expressed in the host cell
for expression of the entire immunoglobulin molecule, as detailed below.
[0212] A variety of host-expression vector systems may be utilized to
express
the antibody molecules of the invention. Such host-expression systems
represent vehicles by which the coding sequences of interest may be produced
and subsequently purified, but also represent cells which may, when
transformed or transfected with the appropriate nucleotide coding sequences,
express an antibody molecule of the invention in situ. These include but are

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
82
not limited to microorganisms such as bacteria (e.g., E. coli, B. subtilis)
transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid
DNA expression vectors containing antibody coding sequences; yeast (e.g.,
Saccharomyces, Pichia) transformed with recombinant yeast expression
vectors containing antibody coding sequences; insect cell systems infected
with recombinant virus expression vectors (e.g., baculoviru.$) containing
antibody coding sequences; plant cell systems infected with recombinant virus
expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic
virus, TMV) or transformed with recombinant plasmid expression vectors
(e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell
systems (e.g., COS, CHO, MK, 293, 3T3 cells) harboring recombinant
expression constructs containing promoters derived from the genome of
mammalian cells (e.g., metallothionein promoter) or from mammalian viruses
(e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter).
Preferably, bacterial cells such as Escherichia coli, and more preferably,
eukaryotic cells, especially for the expression of whole recombinant antibody
molecule, are used for the expression of a recombinant antibody molecule. For
example, mammalian cells such as Chinese hamster ovary cells (CHO), in
conjunction with a vector such as the major intermediate early gene promoter
element from human cytomegalovirus is an effective expression system for
antibodies (Foecking et al., Gene 45:101 (1986); Cockett et al.,
Bio/Technology 8:2 (1990)).
[0213] In bacterial systems, a number of expression vectors may be
advantageously selected depending upon the use intended for the antibody
molecule being expressed. For example, when a large quantity of such a
protein is to be produced, for the generation of pharmaceutical compositions
of
an antibody molecule, vectors which direct the expression of high levels of
fusion protein products that are readily purified may be desirable. Such
vectors
include, but are not limited, to the E. coli expression vector pLTR278 (Ruther
et
al., EMBO J. 2:1791 (1983)), in which the antibody coding sequence may be
ligated individually into the vector in frame with the lac Z coding region so

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
83
that a fusion protein is produced; plN vectors (Inouye & Inouye, Nucleic Acids

Res. 13:3101-3109 (1985); Van Heeke & Schuster, J. Biol. Chem. 24:5503-
5509 (1989)); and the like. pGEX vectors may also be used to express foreign
polypeptides as fusion proteins with glutathione S-transferase (GST). In
general, such fusion proteins are soluble and can easily be purified from
lysed
cells by adsorption and binding to matrix glutathione-agarose beads followed
by elution in the presence of free glutathione. The pGEX vectors are designed
to include thrombin or factor Xa protease cleavage sites so that the cloned
target gene product can be released from the GST moiety.
[0214] In an insect system, Autographa califomica nuclear polyhedrosis
virus
(AcNPV) is used as a vector to express foreign genes. The virus grows in
Spodoptera frugiperda cells. The antibody coding sequence may be cloned
individually into non-essential regions (for example the polyhedrin gene) of
the virus and placed under control of an AcNPV promoter (for example the
polyhalrin promoter).
[0215] In mammalian host cells, a number of viral-based expression systems
may be utilized. In cases where an adenovirus is used as an expression vector,

the antibody coding sequence of interest may be ligated to an adenovirus
transcription/translation control complex, e.g., the late promoter and
tripartite
leader sequence. This chimeric gene may then be inserted in the adenovirus
genome by in vitro or in vivo recombination. Insertion in a non- essential
region of the viral genome (e.g., region El or E3) will result in a
recombinant
virus that is viable and capable of expressing the antibody molecule in
infected
hosts. (e.g., see Logan & Shenk, Proc. Natl. Acad. Sci. USA 81:355-359
(1984)). Specific initiation signals may also be required for efficient
translation of inserted antibody coding sequences. These signals include the
ATG initiation codon and adjacent sequences. Furthermore, the initiation
codon must be in phase with the reading frame of the desired coding sequence
to ensure translation of the entire insert. These exogenous translational
control
signals and initiation codons can be of a variety of origins, both natural and

synthetic. The efficiency of expression may be enhanced by the inclusion of

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
84
appropriate transcription enhancer elements, transcription terminators, etc.
(see
Bittner et al., Methods in Enzymol. 153:51-544 (1987)).
[0216] In addition, a host cell strain may be chosen which modulates the
expression of the inserted sequences, or modifies and processes the gene
product in the specific fashion desired. Such modifications (e.g.,
glycosylation) and processing (e.g., cleavage) of protein products may be
important for the function of the protein. Different host cells have
characteristic and specific mechanisms for the post-translational processing
and modification of proteins and gene products. Appropriate cell lines or host

systems can be chosen to ensure the correct modification and processing of the

foreign protein expressed. To this end, eukaryotic host cells which possess
the
cellular machinery for proper processing of the primary transcript,
glycosylation, and phosphorylation of the gene product may be used. Such
mammalian host cells include but are not limited to CHO, VERY, BHK, Hela,
COS, MDCK, 293, 3T3, W138, and in particular, breast cancer cell lines such
as, for example, BT483, Hs578T, HTB2, BT20 and T47D, and normal
mammary gland cell line such as, for example, CRL7030 and Hs578Bst.
[0217] For long-term, high-yield production of recombinant proteins,
stable
expression is preferred. For example, cell lines which stably express the
antibody molecule may be engineered. Rather than using expression vectors
which contain viral origins of replication, host cells can be transformed with

DNA controlled by appropriate expression control elements (e.g., promoter,
enhancer, sequences, transcription terminators, polyadenylation sites, etc.),
and
a selectable marker. Following the introduction of the foreign DNA,
engineered cells may be allowed to grow for 1-2 days in an enriched media,
and then are switched to a selective media. The selectable marker in the
recombinant plasmid confers resistance to the selection and allows cells to
stably integrate the plasmid into their chromosomes and grow to form foci
which in turn can be cloned and expanded into cell lines. This method may
advantageously be used to engineer cell lines which express the antibody
molecule. Such engineered cell lines may be particularly useful in screening

CA 02548180 2011-12-28
and evaluation of compounds that interact directly or indirectly with the
antibody molecule.
[0218] As described above, a preferred method for producing monoclonal
antibodies is disclosed in US 2002 0123057 Al.
[0219] A number of selection systems may be used, including but not limited
to the herpes simplex virus thymidine kinase (Wigler et al., Cell 11:223
(1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybalska &
Szybalski, Proc. Natl. Acad. Sci. USA 48:202 (1992)), and adenine
phosphoribosyltransferase (Lowy et al., Cell 22:817 (1980)) genes can be
employed in tk-, hgprt- or aprt- cells, respectively. Also, antimetabolite
resistance can be used as the basis of selection for the following genes: dhfi-
,
which confers resistance to methotrexate (Wigler et al., Natl. Acad. Sci. USA
77:357 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt,

which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl.
Acad. Sci. USA 78:2072 (1981)); neo, which confers resistance to the
aminoglycoside G-418 Clinical Pharmacy 12:488-505; Wu and Wu,
Biotherapy 3:87-95 (1991); Tolstoshev, Ann. Rev. Pharmacol. Toxicol.
32:573-596 (1993); Mulligan, Science 260:926-932 (1993); and Morgan and
Anderson, Ann. Rev. Biochem. 62:191-217 (1993); May, 1993, TIB TECH
11(5):155-215); and hygro, which confers resistance to hygromycin (Santerre
et al., Gene 30:147 (1984)). Methods commonly known in the art of
recombinant DNA technology may be routinely applied to select the desired
recombinant clone, and such methods are described, for example, in Ausubel
et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY
(1993); Kriegler, Gene Transfer and Expression, A Laboratory Manual,
Stockton Press, NY (1990); and in Chapters 12 and 13, Dracopoli et al. (eds),
Current Protocols in Human Genetics, John Wiley & Sons, NY (1994);
Colberre-Garapin et al., J. Mol. Biol. 150:1 (1981).
[0220] The expression levels of an antibody molecule can be increased by
vector amplification (for a review, see Bebbington and Hentschel, The use of

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
86
vectors based on gene amplification for the expression of cloned genes in
mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
When a marker in the vector system expressing antibody is amplifiable,
increase in the level of inhibitor present in culture of host cell will
increase the
number of copies of the marker gene. Since the amplified region is associated
with the antibody gene, production of the antibody will also increase (Crouse
et al., MoL Cell. Biol. 3:257 (1983)).
[0221] The host cell may be co-transfected with two expression vectors of
the
invention, the first vector encoding a heavy chain derived polypeptide and the

second vector encoding a light chain derived polypeptide. The two vectors
may contain identical selectable markers which enable equal expression of
heavy and light chain polypeptides. Alternatively, a single vector may be used

which encodes, and is capable of expressing, both heavy and light chain
polypeptides. In such situations, the light chain should be placed before the
heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature
322:52 (1986); Kohler, Proc. Natl. Acad. Sci. USA 77:2197 (1980)). The
coding sequences for the heavy and light chains may comprise cDNA or
genomic DNA.
[0222] Antibodies can be developed against the entire C35 polypeptide or
portions thereof, or sites that are prenylated, glycosylated, phosphorylated,
myristoylated, or amidated.
[0223] Fragments which function as epitopes may be produced by any
conventional means. (See, e.g., Houghten, Proc. NatL Acad. Sci. USA
82:5131-5135 (1985), further described in U.S. Patent No. 4,631,211).
[0224] Similarly, immunogenic epitopes can be used to induce antibodies
according to methods well known in the art. (See, for instance, Sutcliffe et
al.,
supra; Wilson et al., supra; Chow et al., Proc. NatL Acad. Sci. USA 82:910-
914; and Bittle et al., J. Gen. ViroL 66:2347-2354 (1985). Preferred
immunogenic epitopes include the immunogenic epitopes disclosed herein, as
well as any combination of two, three, four, five or more of these
immunogenic epitopes. The polypeptides comprising one or more

CA 02548180 2011-12-28
87
immunogenic epitopes may be presented for eliciting an antibody response
together with a carrier protein, such as an albumin, to an animal system (such

as rabbit or mouse), or, if the polypeptide is of sufficient length (at least
about
25 amino acids), the polypeptide may be presented without a carrier.
However, immunogenic epitopes comprising as few as 8 to 10 amino acids
have been shown to be sufficient to raise antibodies capable of binding to, at

the very least, linear epitopes in a denatured polypeptide (e.g., in Western
blotting). In a specific embodiment, epitopes of the present invention include

those set forth in Table 11, infra.
[0225] Epitope-bearing polypeptides of the present invention may be used to
induce antibodies according to methods well known in the art including, but
not limited to, in vivo immunization, in vitro immunization, and phage
display methods. See, e.g., Sutcliffe et al., supra; Wilson et al., supra, and

Bittle et al., J. Gen. Virol., 66:2347-2354 (1985). If in vivo irru-nunization
is
used, animals may be immunized with free peptide; however, anti-peptide
antibody titer may be boosted by coupling the peptide to a macromolecular
carrier, such as keyhole limpet hemacyanin (KLH) or tetanus toxoid. For
instance, peptides containing cysteine residues may be coupled to a carrier
using a linker such as maleimidobenzoyl- N-hydroxysuccinimide ester (MBS),
while other peptides may be coupled to carriers using a more general linking
agent such as glutaraldehyde.
[0226] Epitope bearing peptides of the invention may also be synthesized as
multiple antigen peptides (MAPs), first described by J. P. Tam in Proc. Natl.
Acad. Sci. U.S.A. 85:5409.
MAPs consist of multiple copies of a specific peptide attached to a
non-immunogenic lysine core. Map peptides usually contain four or eight
copies of the peptide often referred to as MAP-4 or MAP-8 peptides. By way
of non-limiting example, MAPs may be synthesized onto a lysine core matrix
attached to a polyethylene glycol-polystyrene (PEG-PS) support. The peptide
of interest is synthesized onto the lysine residues using 9-
fluorenylmethoxycarbonyl (Fmoc) chemistry. For example, Applied

CA 02548180 2011-12-28
A
88
Biosystems (Foster City, CA) offers MAP resins, such as, for example, the
Fmoc Resin 4 Branch and the Fmoc Resin 8 Branch which can be used to
synthesize MAPs. Cleavage of MAPs from the resin is performed with
standard trifloroacetic acid (TFA)-based cocktails known in the art.
Purification of MAPs, except for desalting, is not necessary. MAP peptides
may be used as an immunizing vaccine which elicits antibodies that recognize
both the MAP and the native protein from which the peptide was derived.
[0227] Epitope bearing peptides of the invention may also be
incorporated into
a coat protein of a virus which can then be used as an immunogen or a vaccine
with which to immunize animals, including humans, in order encourage the
production of anti-epitope antibodies. For example, the V3 loop of the gp120
glycoprotein of the human immunodeficiency virus type 1 (HIV-1) has been
engineered to be expressed on the surface of rhinovirus. Immunization with
this rhinovirus displaying the V3 loop peptide yielded apparently effective
mimics of the HIV-I immunogens (as measured by their ability to be
neutralized by anti-HIV-1 antibodies as well as their ability to elicit the
production of antibodies capable of neutralizing H1V-1 in cell culture). This
techniques of using engineered viral particles as an immunogen is described in

more detail in Smith et al., Behring Inst Mitt Feb;(98):229-39 (1997), Smith
et al, J Virol 72:651-9 (1998), and Zhang etal., Biol Chem 380:365-74 (1999).
[0228] Animals such as rabbits, rats and mice are
immunized with either free
or carrier- coupled peptides or MAP peptides, for instance, by intraperitoneal

and/or intradermal injection of emulsions containing about 100 i_tg of peptide

or carrier protein and Freund's adjuvant or any other adjuvant known for
stimulating an immune response. Several booster injections may be needed,
for instance, at intervals of about two weeks, to provide a useful titer of
anti-
peptide antibody which can be detected, for example, by ELISA assay using
free peptide adsorbed to a solid surface. The titer of anti-peptide antibodies
in
serum from an immunized animal may be increased by selection of anti-
peptide antibodies, for instance, by adsorption to the peptide on a solid
support

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
89
and elution of the selected antibodies according to methods well known in the
art.
[02291 Once an antibody molecule of the invention has been produced by an
animal, chemically synthesized, or recombinantly expressed, it may be purified

by any method known in the art for purification of an immunoglobulin
molecule, for example, by chromatography (e.g., ion exchange, affinity,
particularly by affinity for the specific antigen after Protein A, and sizing
column chromatography), centrifugation, differential solubility, or by any
other
standard technique for the purification of proteins. In addition, the
antibodies
of the present invention or fragments thereof can be fused to heterologous
polypeptide sequences described herein or otherwise known in the art, to
facilitate purification.
[0230) The antibodies of the present invention may be generated by any
suitable method known in the art. Polyclonal antibodies to an antigen-of-
interest can be produced by various procedures well known in the art. For
example, a polypeptide of the invention can be administered to various host
animals including, but not limited to, rabbits, mice, rats, etc. to induce the

production of sera containing polyclonal antibodies specific for the antigen.
Various adjuvants may be used to increase the immunological response,
depending on the host species, and include but are not limited to, Freund's
(complete and incomplete), mineral gels such as aluminum hydroxide, surface
active substances such as lysolecithin, pluronic polyols, polyanions,
peptides,
oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially
useful human adjuvants such as BCG (bacille Calmette-Guerin) and
corynebacterium parvum. Such adjuvants are also well known in the art.
[0231] Monoclonal antibodies can be prepared using a wide variety of
techniques known in the art including the use of hybridoma, recombinant, and
phage display technologies, or a combination thereof. For example,
monoclonal antibodies can be produced using hybridoma techniques including
those known in the art and taught, for example, in Harlow et al., Antibodies:
A
Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988);

CA 02548180 2011-12-28
Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-
681 (Elsevier, N.Y., 1981).
The term "monoclonal antibody" as used herein is not limited to
antibodies produced through hybridoma technology. The term "monoclonal
antibody" refers to an antibody that is derived from a single clone, including

any eukaryotic, prokaryotic, or phage clone, and not the method by which it is

produced.
[0232] Methods for producing and screening for specific antibodies using
hybridoma technology are routine and well known in the art and are discussed
in detail in the Examples. In a non-limiting example, mice can be immunized
with a polypeptide of the invention or a cell expressing such peptide. Once an

immune response is detected, e.g., antibodies specific for the antigen are
detected in the mouse serum, the mouse spleen is harvested and splenocytes
isolated. The splenocytes are then fused by well known techniques to any
suitable myeloma cells, for example cells from cell line SP20 available from
the ATCC. Hybridomas are selected and cloned by limited dilution. The
hybridoma clones are then assayed by methods known in the art for cells that
secrete antibodies capable of binding a polypeptide of the invention. Ascites
fluid, which generally contains high levels of antibodies, can be generated by

immunizing mice with positive hybridoma clones.
[0233] Accordingly, the present invention provides methods of generating
monoclonal antibodies as well as antibodies produced by the method
comprising culturing a hybridoma cell secreting an antibody of the invention
wherein, preferably, the hybridoma is generated by fusing splenocytes isolated

from a mouse immunized with an antigen of the invention with myeloma cells
and then screening the hybridomas resulting from the fusion for hybridoma
clones that secrete an antibody able to bind a polypeptide of the invention.
[02341 Antibody fragments which recognize specific epitopes may be
generated by known techniques. For example, Fab and F(ab')2 fragments of the
invention may be produced by proteolytic cleavage of immunoglobulin
molecules, using enzymes such as papain (to produce Fab fragments) or pepsin

CA 02548180 2011-12-28
91
(to produce F(ab')2 fragments). F(ab')2 fragments contain the variable region,

the light chain constant region and the CHI domain of the heavy chain.
[0235] For example, the antibodies of the present invention can also be
generated using various phage display methods known in the art. In phage
display methods, functional antibody domains are displayed on the surface of
phage particles which carry the polynucleotide sequences encoding them. In a
particular embodiment, such phage can be utilized to display antigen binding
domains expressed from a repertoire or combinatorial antibody library (e.g.,
human or murine). Phage expressing an antigen binding domain that binds the
antigen of interest can be selected or identified with antigen, e.g., using
labeled
antigen or antigen bound or captured to a solid surface or bead. Phage used in

these methods are typically filamentous phage including fd and MI3 binding
domains expressed from phage with Fab, Fv or disulfide stabilized Fv
antibody domains recombinantly fused to either the phage gene m or gene VIII
protein. Examples of phage display methods that can be used to make the
antibodies of the present invention include those disclosed in Brinkman et
al.,
J. Immunol. Methods /82:41-50 (1995); Ames et al., J. Immtwol. Methods
/84:177-186 (1995); Kettleborough et al., Eur. J. Immunol. 24:952-958
(1994); Persic et al., Gene 187 9-18 (1997); Burton et al., Advances in
Immunology 57:191-280 (1994); PCT application No. PCT/GB91/01134; PCT
publications WO 90/02809; WO 91/10737; WO 92/01047; WO 92/18619;
WO 93/11236; WO 95/15982; WO 95/20401; and U.S. Pat. Nos. 5,698,426;
5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047; 5,571,698;
5,427,908; 5,516,637; 5,780,225; 5,658,727; 5,733,743 and 5,969,108.
[0236] As described in the above references, after phage selection, the
antibody coding regions from the phage can be isolated and used to generate
whole antibodies, including human antibodies, or any other desired antigen
binding fragment, and expressed in any desired host, including mammalian
cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in
detail
below. For example, techniques to recombinantly produce Fab, Fab' and

CA 02548180 2011-12-28
92
F(ab')2 fragments can also be employed using methods known in the art such
as those disclosed in PCT publication WO 92/22324; Mullinax et al.,
BioTechniques /2(6):864-869 (1992); and Sawai et al., AJRI 34:26-34 (1995);
and Better et al., Science 240:1041-1043 (1988).
[0237] Examples of
techniques which can be used to produce single-chain Fvs
and antibodies, as well as diabodies, triabodies, and tetrabodies, include
those
described in U.S. Pat. Nos. 4,946,778 and 5,258,498; Huston et al., Methods in

Enzymology 203:46-88 (1991); Shu et al., PNAS 90:7995-7999 (1993); Skerra
et al., Science 240:1038-1040 (1988); U.S. Publication No. 20020018749 and
U.S. Pat. No. 5,837,242. For some uses, including in vivo use of antibodies in

humans and in vitro detection assays, it may be preferable to use chimeric,
humanized, or human antibodies. A chimeric antibody is a molecule in which
different portions of the antibody are derived from different animal species,
such as antibodies having a variable region derived from a murine monoclonal
antibody and a human immunoglobulin constant region. Methods for
producing chimeric antibodies are known in the art. See e.g., Morrison,
Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et
al.,
J. Immunol. Methods /25:191-202 (1989); U.S. Pat. Nos. 5,807,715;
4,816,567; and 4,816397.
Humanized antibodies are antibody molecules from non-human
species antibody that binds the desired antigen having one or more
complementarity determining regions (CDRs) from the non-human species
and a framework regions from a human immunoglobulin molecule. Often,
framework residues in the human framework regions will be substituted with
the corresponding residue from the CDR donor antibody to alter, preferably
improve, antigen binding. These framework substitutions are identified by
methods well known in the art, e.g., by modeling of the interactions of the
CDR and framework residues to identify framework residues important for
antigen binding and sequence comparison to identify unusual framework
residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No.

CA 02548180 2011-12-28
93
5,585,089; Riechmann et al., Nature 332:323 (1988)).
Antibodies can be humanized using a
variety of techniques known in the art including, for example, CDR-grafting
(EP 239,400; PCT publication WO 91/09967; U.S. Pat. Nos. 5,225,539;
5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596;
Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al.,
Protein Engineering 7(6):805-814 (1994); Roguska. et al., PNAS 91:969-973
(1994)), and chain shuffling (U.S. Pat. No. 5,565,332).
[0238] Completely human antibodies are particularly desirable for
therapeutic
treatment of human patients. Human antibodies can be made by a variety of
methods known in the art including phage display methods described above
using antibody libraries derived from human immunoglobulin sequences, and
the methods disclosed in US 2002 0123057 Al. See also, U.S. Pat. Nos.
4,444,887 and 4,716,111; and PCT publications WO 98/46645, WO 98/50433,
WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO
91/10741.
[0239] Human antibodies can also be produced using transgenic mice which
are incapable of expressing functional endogenous immunoglobulins, but
which can express human immunoglobulin genes. For example, the human
heavy and light chain immunoglobulin gene complexes may be introduced
randomly or by homologous recombination into mouse embryonic stem cells.
Alternatively, the human variable region, constant region, and diversity
region
may be introduced into mouse embryonic stem cells in addition to the human
heavy and light chain genes. The mouse heavy and light chain
immunoglobulin genes may be rendered non-functional separately or
simultaneously with the introduction of human immunoglobulin loci by
homologous recombination. In particular, homozygous deletion of the JH
region prevents endogenous antibody production. The modified embryonic
stem cells are expanded and microinjected into blastocysts to produce chimeric

mice. The chimeric mice are then bred to produce homozygous offspring
which express human antibodies. The transgenic mice are immunized in the

CA 02548180 2011-12-28
94
normal fashion with a selected antigen, e.g., all or a portion of a
polypeptide of
the invention. Monoclonal antibodies directed against the antigen can be
obtained from the immunized, transgenic mice using conventional hybridoma
technology. The human immunoglobulin transgenes harbored by the transgenic
mice rearrange during B cell differentiation, and subsequently undergo class
switching and somatic mutation. Thus, using such a technique, it is possible
to
produce therapeutically useful IgG, IgA, IgM and IgE antibodies. For an
overview of this technology for producing human antibodies, see Lonberg and
Huszar, Int. Rev. hnmunol. 13:65-93 (1995). For a detailed discussion of this
technology for producing human antibodies and human monoclonal antibodies
and protocols for producing such antibodies, see, e.g., PCT publications WO
98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European Patent No.
0 598 877; U.S. Pat. Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825;
5,661,016; 5,545,806; 5,814,318; 5,885,793; 5,916,771; and 5,939,598.
In addition, companies
such as Abgenix, Inc. (Freemont, Calif.) and Genpharm (San Jose, Calif.) can
be engaged to provide human antibodies directed against a selected antigen
using technology similar to that described above.
[0240] Completely human antibodies which recognize a selected epitope can
be generated using a technique referred to as "guided selection." In this
approach a selected non-human monoclonal antibody, e.g., a mouse antibody,
is used to guide the selection of a completely human antibody recognizing the
same epitope. (Jespers et al., Bio/technology /2:899-903 (1988)).
[0241] Further, antibodies to the polypeptides of the invention can, in
turn, be
utilized to generate anti-idiotype antibodies that "mimic" polypeptides of the

invention using techniques well known to those skilled in the art. (See, e.g.,

Greenspan & Bona, FASEB J. 7(5):437-444; (1989) and Nissinoff, J. Imrnunol.
147(8):2429-2438 (1991)). For example, antibodies which bind to and
competitively inhibit polypeptide multimerization and/or binding of a
polypeptide of the invention to a ligand can be used to generate anti-
idiotypes
that "mimic" the polypeptide multimerization and/or binding domain and, as a

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
consequence, bind to and neutralize polypeptide and/or its ligand. Such
neutralizing anti-idiotypes or Fab fragments of such anti-idiotypes can be
used
in therapeutic regimens to neutralize polypeptide ligand. For example, such
anti-idiotypic antibodies can be used to bind a polypeptide of the invention
and/or to bind its ligands/receptors, and thereby block its biological
activity.
Antibody Conjugates
[0242] In another embodiment, the invention provides a method for the
specific destruction of cells (e.g., the destruction of aberrant cells,
including,
but not limited to, tumor cells) by administering antibodies against cancer-
associated, intracellular antigens in association with toxins or cytotoxic
prodrugs.
[0243] By "toxin" is meant compounds that bind and activate endogenous
cytotoxic effector systems, radioisotopes, holotoxins, modified toxins,
catalytic subunits of toxins, cytotoxins (cytotoxic agents), or any molecules
or
enzymes not normally present in or on the surface of a cell that under defined

conditions cause the cell's death. Toxins that may be used according to the
methods of the invention include, but are not limited to, radioisotopes known
in the art, compounds such as, for example, antibodies (or complement fixing
containing portions thereof) that bind an inherent or induced endogenous
cytotoxic effector system, thymidine kinase, endonuclease, RNAse, alpha
toxin, ricin, abrin, Pseudomonas exotoxin A, diphtheria toxin, saporin,
momordin, gelonin, pokeweed antiviral protein, alpha-sarcin and cholera
toxin. "Toxin" also includes a cytostatic or cytocidal agent, a therapeutic
agent
or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi,
or
, ,
other radioisotopes such as, for example, 103pd, 133xe, 1311 68-e 57Co, 65Zn,
85sr, 32p, 35s, 90y, 153sm, 153Gd, 169yb, 51cr, 54mn, 75se, 113sn, 90yttri
117Tin, 186Rhenium, 166Hohnium, and 188Rhenium and those listed in the
"Complex" section below; luminescent labels, such as luminol; and
fluorescent labels, such as fluorescein and rhodamine, and biotin. In a
specific

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
96
embodiment, antibodies of the present invention (including fragments or
variants thereof) are conjugated with a radioisotope, e.g., 131I. In a
preferred
embodiment, the radioisotope-conjugated antibody is adminstered for
radioimmunotherapy in combination with chemotherapy.
[0244] A cytotoxin or cytotoxic agent includes any agent that is
detrimental to
cells. Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium
bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine,
colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione,
mitoxantrone, mithramycin, actinomycin D, 1 -dehydrotesto sterone,
glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin
and analogs or homologs thereof. Cytotoxin or cytotoxic agents include, but
are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-
thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g.,

mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and
lomustine (CCNU), cyclothospharnide, busulfan, dibromomannitol,
streptozotocin, mitomycin C, and cis- dichlorodiamine platinum (II) (DDP)
cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and
doxorubicin), .antibiotics (e.g., dactinomycin (formerly actinomycin),
bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents
(e.g., vincristine and vinblastine).
[0245] By "cytotoxic prodrug" is meant a non-toxic compound that is
converted by an enzyme, normally present in the cell, into a cytotoxic
compound. Cytotoxic prodrugs that may be used according to the methods of
the invention include, but are not limited to, glutamyl derivatives of benzoic

acid mustard alkylating agent, phosphate derivatives of etoposide or
mitomycin C, cytosine arabinoside, daunorubisin, and phenoxyacetamide
derivatives of doxorubicin.
[0246] The present invention also encompasses antibodies recombinantly
fused or chemically conjugated (including both covalently and non-covalently
conjugations) to a polypeptide (or portion thereof, preferably at least 10,
20,
30, 40, 50, 60, 70, 80, 90 or 100 amino acids of the polypeptide) to generate

CA 02548180 2011-12-28
97
fusion proteins. The fusion does not necessarily need to be direct, but may
occur through linker sequences. The antibodies may be specific for antigens
other than C35 polypeptides (or portion thereof, preferably at least 10, 20,
30,
40, 50, 60, 70, 80, 90 or 100 amino acids of the polypeptide). In particular,
antibodies may be specific for other cancer-associated, intracellular
antigens.
In a preferred embodiment, such intracellular antigens are prenylated
proteins.
[0247] Antibodies of the present invention (including fragments or variants
thereof) may be fused to either the N- or C-terminal end of the heterologous
protein (e.g., immtmoglobulin Fc polypeptide or human serum albumin
polypeptide). Antibodies of the invention may also be fused to albumin
(including but not limited to recombinant human serum albumin (see, e.g.,
U.S. Patent No. 5,876,969, issued March 2, 1999, EP Patent 0 413 622, and
U.S. Patent No. 5,766,883, issued June 16, 1998)),
resulting in chimeric polypeptides. In a preferred
embodiment, antibodies of the present invention (including fragments or
variants thereof) are fused with the mature form of human serum albumin (i.e.,

amino acids 1 ¨ 585 of human serum albumin as shown in Figures 1 and 2 of
EP Patent 0 322 094).
In another preferred embodiment, antibodies of the present invention
(including fragments or variants thereof) are fused with polypeptide fragments

comprising, or alternatively consisting of, amino acid residues 1-z of human
serum albumin, where z is an integer from 369 to 419, as described in U.S.
Patent 5,766,883.
[0248] Polynucleotides encoding fusion proteins of the invention are also
encompassed by the invention. Such fusion proteins may, for example,
facilitate purification and may increase half-life in vivo. Antibodies fused
or
conjugated to polypeptides may also be used in in vitro immunoassays and
purification methods using methods known in the art. See e.g., Harbor et al.,
supra, and PCT publication WO 93/21232; EP 439,095; Naramura et al.,
Immunol. Lett. 39:91-99 (1994); U.S. Patent 5,474,981; Gullies et al., PNAS

CA 02548180 2011-12-28
98
89:1428-1432 (1992); Fell et aL, J. Imrnunol. /46:2446-2452(1991).
[0249] Moreover, the antibodies or fragments thereof of the present
invention
can be fused to marker sequences, such as a peptide to facilitate
purification.
In preferred embodiments, the marker amino acid sequence is a hexa-histidine
peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton
Avenue, Chatsworth, Calif., 91311), among others, many of which are
commercially available. As described in Gentz et al., Proc. Natl. Acad. Sci.
USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient
purification of the fusion protein. Other peptide tags useful for purification

include, but are not limited to, the "HA" tag, which corresponds to an epitope

derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767
(1984)) and the "flag" tag.
[02501 The present invention further encompasses antibodies, such as C35
antibodies or fragments thereof, conjugated to a diagnostic or therapeutic
agent. The antibodies can be used diagnostically to, for example, monitor the
development or progression of a tumor as part of a clinical testing procedure
to, e.g., determine the efficacy of a given treatment regimen. Detection can
be
facilitated by coupling the antibody to a detectable substance. Examples of
detectable substances include various enzymes, prosthetic groups, fluorescent
materials, luminescent materials, bioluminescent materials, radioactive
materials, positron emitting metals using various positron emission
tomographies, and nonradioactive paramagnetic metal ions. The detectable
substance may be coupled or conjugated either directly to the antibody (or
fragment thereof) or indirectly, through an intermediate (such as, for
example,
a linker known in the art) using techniques known in the art. See, for
example,
U.S. Pat. No. 4,741,900 for metal ions which can be conjugated to antibodies
for use as diagnostics according to the present invention. Examples of
suitable
enzymes include horseradish peroxidase, alkaline phosphatase, beta-
galactosidase, or acetylcholinesterase; examples of suitable prosthetic group
complexes include streptavidin/biotin and avidin/biotin; examples of suitable

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
99
fluorescent materials include umbelliferone, fluorescein, fluorescein
isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride

or phycoerythrin; an example of a luminescent material includes luminol;
examples of bioluminescent materials include luciferase, luciferin, and
aequorin; and examples of suitable radioactive material include 1251, 1311,
111In or 99Tc.
[0251] The therapeutic agent is not to be construed as limited to
classical
chemical therapeutic agents. For example, the therapeutic agent may be a
protein or polypeptide possessing a desired biological activity. Such proteins

may include, for example, a toxin such as abrin, ricin A, pseudomonas
exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha-
interferon, beta-interferon, nerve growth factor, platelet derived growth
factor,
tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta,
AIM I (See, International Publication No. WO 97/33899), AIM 11 (See,
International Publication No. WO 97134911), Fas Ligand (Takahashi et al.,
Int. Immunol., 6:1567-1574 (1994)), VEGI (See, International Publication No.
WO 99/23105), a thrombotic agent or an anti- angiogenic agent, e.g.,
angiostatin or endostatin; or, biological response modifiers such as, for
example, lymphokines, interleukin-1 ("IL-1"), interleukin-2 ("IL-2"),
interleukin-6 ("IL-6"), granulocyte macrophage colony stimulating factor
("GM-CSF"), granulocyte colony stimulating factor ("G-CSF"), or other
growth factors.
[0252] Antibodies may also be attached to solid supports, which are
particularly useful for immunoassays or purification of the target antigen.
Such
solid supports include, but are not limited to, glass, cellulose,
polyacrylamide,
nylon, polystyrene, polyvinyl chloride or polypropylene.
[0253] Techniques for conjugating such toxins and therapeutic agents to
antibodies are well known, see, e.g., Arnon et al., "Monoclonal Antibodies For

Immunotargeting Of Drugs In Cancer Therapy", in Monoclonal Antibodies
And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc.
1985); Hellstrom et al., "Antibodies For Drug Delivery", in Controlled Drug

CA 02548180 2011-12-28
100
Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc.
1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A
Review", in Monoclonal Antibodies '84: Biological And Clinical Applications,
Pinchera et al. (eds.), pp. 475-506 (1985); "Analysis, Results, And Future
Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer
Therapy", in Monoclonal Antibodies For Cancer Detection And Therapy,
Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al.,
"The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates",
Immunol. Rev. 62:119-58 (1982).
[0254] Alternatively, an antibody can be conjugated to a second antibody to
form an antibody heteroconjugate as described by Segal in U.S. Pat. No.
4,676,980.
[0255] An antibody, with or without a toxin or therapeutic agent conjugated
to
it, administered alone or in combination with cytotoxic factor(s) and/or
cytolcine(s) can be used as a therapeutic.
[0256] Techniques known in the art may be applied to label antibodies of
the
invention. Such techniques include, but are not limited to, the use of
bifunctional conjugating agents (see e.g., U.S. Pat. Nos. 5,756,065;
5,714,631;
5,696,239; 5,652,361; 5,505,931; 5,489,425; 5,435,990; 5,428,139; 5,342,604;
5,274,119; 4,994,560; and 5,808,003).
[0257] Chelator molecules are known in the art. For example, see
Subrarnanian, R. and Meares, C.F., "Bifunctional Chelating Agents for
Radiometal-labeled monoclonal Antibodies," in Cancer Imaging with
Radiolabeled Antibodies (D. M. Goldenberg, Ed.) Kluwer Academic
Publications, Boston; Saji, H., "Targeted delivery of radiolabeled imaging and

therapeutic agents: bifunctional radiopharmaceuticals." Crit. Rev. Ther. Drug
Carrier Syst. 16:209-244 (1999); Srivastava S.C. and Mease R.C., "Progress in
research on ligands, nuclides and techniques for labeling monoclonal
antibodies." Int. J. Rad. Appl. Instrum. B 18:589-603 (1991); and Liu, S. and
Edwards, D.S., "Bifunctional chelators for therapeutic lanthanide

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
101
radiopharmaceuticals." Bioconjug. Chem. 12:7-34 (2001). Any chelator which
can be covalently bound to said antibody may be used according to the present
invention. The chelator may further comprise a linker moiety that connects the

chelating moiety to the antibody.
[0258] Bifunctional chelators based on macrocyclic ligands in which
conjugation is via an activated arm attached to the carbon backbone of the
ligand can be employed as described by M. Moi et al., J. Amer. Chem. Soc.
49:2639 (1989) (2-p-nitrobenzy1-1,4,7,10-tetra a 7acyclodo decane-N,N,N",1\l'
" -
tetraacetic acid); S. V. Deshpande et al., J. NucL Med. 31:473 (1990); G.
Ruser et al., Bioconj. Chem. /:345 (1990); C. J. Broan et al., J. C. S. Chem.
Comm. 23:1739 (1990); and C. J. Anderson et al., J. NucL Med. 36:850
(1995).
[0259] In one embodiment, the chelator is a macrocyclic chelator, such
as
polyazamacrocyclic chelators, optionally containing one or more carboxy,
phosphonate, or phosphate groups. In another embodiment, the chelator is a
chelator selected from the group consisting of DOTA, analogues of DOTA,
and derivatives of DOTA.
[0260] In one
embodiment, suitable chelator molecules include DOXA (1-
oxa-4,7,10-triazacyclodo decanetriacetic acid), NOTA (1,4,7-
triazacyclononanetriacetic acid), TETA (1,4,8,11-

tetraazacyclotetradecanetetraacetic acid), and THT (4'-(3-amino-4-methoxy-
pheny1)-6,6"-bis(N',N'-dicarboxymethyl-N-methylhydra zino)-
2,2':6',2"-
terpyridine), and analogs and derivatives thereof. See, e.g., Ohmono et al.,
J.
Med. Chem. 35: 157-162 (1992); Kung et al., J. NucL Med. 25: 326-332
(1984); Jurisson et al., Chem. Rev. 93:1137-1156 (1993); and U.S. Patent No.
5,367,080. Other suitable chelators include chelating agents disclosed in U.S.

Patent Nos. 4,647,447; 4,687,659; 4,885,363; EP-A-71564; W089/00557; and
EP-A-232751.
[0261] In another embodiment, suitable macrocyclic carboxylic acid
chelators
which can be used in the present invention include 1,4,7,10-
tetraazacyclododecane-N,N',N",Nm-tetraacetic acid (DOTA); 1,4,8,12-

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
102
tetraazacyclop entadecane-N,N',N",N' "-tetraacetic acid (15N4);
1,4,7-
triazacyclononane-N,N',N"-triacetic acid (9N3); 1,5,9-triazacyclododecane-
N,N',N"-triacetic acid (12N3); and 6-bromoacetamido-benzy1-1,4,8,11-
tetraazacyclotetadecane- N,NI,N",Nm-tetraacetic acid (BAT).
[0262] The antibody conjugate may contain more than an average of one
chelator molecule. In one embodiment, the antibody conjugate contains about
1, 2, 3, 4, 5, 6, 7, 8,9, 10, 15, 20, 25, 30, or 35 chelator molecules. In
another
embodiment, the antibody conjugate contains about 1, 2, 3, 4, 5, 6, 7, 8, 9,
or
chelator molecules. In another embodiment, the antibody conjugate
contains about 1, 2, 3, 4, or 5 chelator molecules. In another embodiment, the

antibody conjugate contains an average of about 1 chelator molecule.
[0263] The antibody conjugate, as described above, comprises an
antibody and
a chelator molecule covalently bonded together. The covalent bond between
the protein and chelator is formed between an atom of the protein and an atom
of the chelator. The atom of the protein that forms the covalent bond can be
any suitable atom of the protein, such as a carbon, nitrogen, oxygen, and
sulfur. Certain functional groups on the protein are preferred to form the
covalent bond with the chelator. Such groups include an amino group at the
terminus of the protein; a carboxy group at the terminus of the protein; an
amino group on a lysine residue; a carboxy group on an aspartame or
glutamate residue; a guanidino group on an arginine residue; a thiol group on
a
cysteine residue; a hydroxy group on a serine residue or tlareonine residue; a

imidazole group of a histidine residue; a hydroxy group of a tyrosine; a
hydroxy group of a tryptophan group; and an amide group of an asp aragine or
glutamine residue.
Methods Of Preparing Antibody Conjugates
[0264] An
additional aspect of the present invention is directed to a method of
preparing an antibody conjugate. The general procedure for preparing an
antibody conjugate comprises reacting an antibody with a chelator. A suitable

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
103
chelator will be able to react with the antibody to form a covalent bond
between the chelator and the antibody.
[0265] A chelator that can form part of the conjugate is known in the art,
and
methods of preparing such a chelator are known. Any chelator which is able
to form a covalent bond with the antibody may be used to form the antibody
conjugate. Such chelators are described below.
[0266] In one embodiment, a chelator to be used in the method according to
the present invention is an activated chelator. The activated chelator
contains
a functional group that will readily react with a functional group on the
protein, thereby forming a covalent bond between the protein and the chelator.

Such functional groups are well known in the art.
[0267] A suitable reactive functional group is a group that will react
directly
with carboxy, aldehyde, amine, alcohol, or sulfhydryl group on the antibody.
Such groups include, for example, active halogen containing groups including,
for example, chloromethylphenyl groups and chloroacetyl (C1CH2C(=0)-)
groups; activated 2-(leaving group substituted)-ethylsulfonyl and
ethylcarbonyl
groups such as 2-chloroethylsulfonyl and 2-chloroethylcarbonyl; vinylsulfonyl;

vinylcarbonyl; epoxy; isocyanato; isothiocyanato; aldehyde; aziridine;
succinimidoxycarbonyl; activated acyl groups such as carboxylic acid halides;
and mixed anhydrides.
[0268] A chelator which can be used in the present method includes any
specific chelator as described herein for the antibody conjugate. Moreover, in

another embodiment, the chelator used in the method of the invention is a
chelator which can be used to prepare any specific antibody conjugate as
described above. In one embodiment, the chelator is an activated chelator
selected from the group consisting of DOTA, analogues of DOTA, and
derivatives of DOTA, wherein each of said DOTA, analogues of DOTA, and
derivatives of DOTA contains a suitable activating group which enables the
chelator molecule to be covalently bonded to the antibody.
[0269] In another embodiment, the activated chelator for use in preparing
a
conjugate according to the present invention is a-(5-isothiocyanato- 2-

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
104
methoxypheny1)-1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid,
which is also known as Me0-DOTA-NCS. A salt or ester of a-(5-
isothiocyanato- 2-methoxypheny1)- 1,4,7,10-tetraazacyclododecane-1,4,7,10-
tetraacetic acid may also be used.
[0270] In one embodiment, the method of preparing an antibody conjugate
comprises mixing, agitating, or preparing a solution, said solution comprising

an antibody and a chelator. In an additional embodiment, the method of
preparing an antibody conjugate comprises mixing, agitating, or preparing a
solution comprising an antibody, a chelator, citrate buffer, HEPES buffer, and

sterile water. In an additional embodiment, the solution further comprises
NaOH. In an additional embodiment, the solution has a pH of about 8.5.
[0271] In an additional embodiment, the method of preparing the
conjugate
comprises a) mixing, agitating, or preparing a solution comprising an antibody

and a chelator, at a temperature of about 0 C to about 50 C for about 0.5
hours to about 24 hours, wherein said solution has a pH of about 8.0 to about
9.0; and b) optionally adding a quenching agent.
[0272] In one embodiment of the above process, the solution is mixed,
agitated or prepared at a temperature of about 0 C to about 50 C. In another

embodiment of the above process, the solution is mixed, agitated or prepared
at a temperature of about 20 C to about 30 C. The temperature at which the
solution is mixed, agitated, or prepared can be about 0, 5, 10, 15, 20, 25,
20,
35, 40, 45, or 50 C.
[0273] Any suitable chelator as described above may be used in the
present
process.
[0274] The molar ratio of antibody to chelator used in the preparation
of the
conjugate can vary. The molar ratio of antibody to chelator can be from about
1000:1 to about 1:1000, from about 1 to about 100, from about 5 to about 20,
from about 10 to about 15, about 12. The molar ratio of antibody to chelator
refers to the ratio of the number of antibody molecules to the number of
chelator molecules in the reaction solution.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
105
[0275] When preparing an antibody conjugate, the solution can be mixed,
agitated, or allowed to stand for a variable number of hours, depending upon a

number of variables, such factors including the identity of the chelator, the
identity of the antibody, the temperature of the reaction, the pH, the
identity of
the buffer, the molar ratio of the reactants, the purity of the available
reagents,
the presence of a catalyst or activator, and other factors which would be
evident to one of skill in the art. In one embodiment of the above process,
the
solution comprising an antibody and a chelator is mixed, agitated, or allowed
to stand for about 0.5 hours to about 24 hours. In another embodiment, the
solution is mixed, agitated, or allowed to stand for about 1 hour to about 20
hours. In another embodiment, the solution is mixed, agitated, or allowed to
stand for about 2 hour to about 10 hours. In another embodiment, the solution
is mixed, agitated, or allowed to stand for about 3 hour to about 5 hours. In
another embodiment, the solution is mixed, agitated, or allowed to stand for
about 4 hours. In other embodiments, the solution is mixed, agitated, or
allowed to stand for about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 ,15,
16, 17,
18, 19, 20, 21, 22, 23, or 24 hours.
[0276] The pH of the solution used to prepare the antibody conjugate can
vary.
In one embodiment, the pH will be about 6, 7, 8, 9, or 10. In another
embodiment, said solution has a pH of about 7 to about 10. In another
embodiment, said solution has a pH of about 7.5 to about 9.5. In another
embodiment, said solution has a pH of about 8 to about 9. In another
embodiment, said solution has a pH of about 8.5.
[0277] The solution used to prepare the antibody conjugate may further
comprise a buffer. Buffers are well-known in the art. Suitable buffers for use

in the preparation of an antibody are described below for the method of
preparing an antibody conjugate. In one embodiment, the buffer is a citrate
buffer or an acetate buffer. In another embodiment, the buffer includes an
acetate buffer having a concentration of about 1 to about 50 mM and having a
NaC1 concentration of about 1 to about 500 mM. In another embodiment, the
buffer includes an acetate buffer having a concentration of about about 10 mM

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
106
and having a NaC1 concentration of about about 140 mM. Suitable acetate
buffers include acetate buffers havaing a concentration of about 1, 5, 10, 15,

20, 25, 20, 35, 40, 45, or 55 mM. Suitable buffers and solutions include those

having a NaC1 concentration of about 1, 50, 75, 100, 125, 140, 150, 175, 200,
225, 250, 275, 300, 350, 400, 450, or 500 mM. An additional suitable buffer
is a HEPES buffer, in particular a HEPES buffer having a concentration of
about 100, 200, 300, 400, 500, 600, 700, 800, 900, or 1000 mM. In an
additional embodiment, the solution comprises a HEPES buffer having a
concentration of about 500 mM.
[0278] The concentration of the antibody used in the method of the
invention
may vary. In one embodiment, the concentration of antibody in the solution is
from about 0.1 mg/mL to about 10 mg/mL. In another embodiment, the
antibody concentration is about 0.5 mg/mL to about 5 mg/mL. In other
specific embodiments, the antibody concentration is about 0.1, 0.2, 0.3, 0.4,
0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9,2.0,
2..1, 2.2,
2.3, 2.4, or 2.5 mg/mL. In another specific embodiment, the concentration of
antibody in the solution is about 1 mg/mL to about 2 mg/mL.
[0279] A reagent which facilitates the formation of the conjugate can also
be
used. Such reagents typically activate a chelator to react more readily with a

protein. Alternatively, the reagent may activate the protein to react more
readily with the chelator. Examples of such reagents are known in the art and
include dicyclohexylcarbodiimide (DCC), diethylazodicarboxylate (DEAD),
and diisopropylazodicarboxylate (DIAD).
[0280] According to the method of the present invention, a quencing agent
may optionally be added to the reaction solution after the antibody conjugate
is
prepared to a sufficient degree. Suitable quencing agents are known in the
art.
In one embodiment, the quenching agent is glycine. In a specific embodiment,
a solution comprising a glycine buffer is used as a quenching example. By
way of example, after the reaction solution comprising the antibody and the
chelator has been mixed, agitated, or allowed to stand for a sufficient amount

of time, e.g., 1-10 hours, 3-5 hours, or 1-3 hours, a solution comprising a

CA 02548180 2006-06-01
WO
2005/055936 PCT/US2004/040573
107
glycine buffer is added to the reaction solution to stop the reaction. The
reaction solution is then allowed to stand for an additional amount of time,
e.g., about 0.25, 0.5, 0.75, 1, or 1.5 hours.
[0281] The method of preparing an antibody conjugate may.comprise mixing,
agitating, or preparing a solution comprising:
an antibody has a concentration of about 1 to about 2 mg/mL;
a Me0-DOTA-NCS in an amount such that the ratio of antibody to
Me0-DOTA-NCS is about 1 to 12;
a citrate buffer having a concentration of about 0.5 to about 20 mM;
NaClhaing a concentration of about 1 to about 200 mM;
HEPES buffer having a concentration of about 10 to about 500 mM;
and
sterile water.
[0282] The method may comprise:
preparing a first solution comprising an antibody and a citrate buffer;
adding a second solution comprising a HEPES buffer to said first
solution;
adding a third solution comprising a chelator and NaOH to said first
solution;
optionally adjusting the pH of said first solution to about 8.5;
mixing, agitating, or allowing to stand said first solution at about 25 C
for about 3 to about 5 hours; and
optionally adding quencing agent to said first solution.
[0283] The method of preparing an antibody conjugate according to the
present invention further optionally comprises a process of purifying said
conjugate. A number of known methods may be used to purifying said protein
conjugate.
[0284] In one embodiment, the purifying step uses normal flow filtration or
tangential flow filtration. In another embodiment, the process for purifying
the
conjugate according to the present invention is a diafiltration method.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
108
Antibody Complexes
[0285] An additional aspect of the present invention is directed to an
antibody
complex comprising an antibody conjugate and a metal ion, wherein said
metal ion is noncovalently associated with the chelator moiety of said
antibody
conjugate. Specific embodiments of an antibody complex of the invention
include a complex comprising an antibody conjugate, as set forth herein, and a

metal ion
[0286] Any metal ion that noncovalently associates with the chelating
moiety
of said antibody conjugate may be used. Such a metal ion includes a metal ion
selected from the group consisting of Ac, Ag, At, Au, Bi, Ce, Co, Cr, Cu, Dy,
Er, Eu, Fe, Ga, Gd, Hg, Ho, In, La, Lu, Mn, Mo, Nd, Ni, Os, Pb, Pd, Pm, Pr,
Pt, Rb, Re, Rh, Ru, Sb, Sc, Si, Sm, Sn, Sr, Tb, Tc, Tl, Tm, V, W, Y, and Yb.
[0287] In one embodiment of the present invention, the metal ion of the
complex is a radionuclide. Radionuclides useful in the antibody complex are
known in the art. The radionuclides useful in the present invention include
gamma-emitters, positron-emitters, x-ray emitters, fluorescence-emitters, beta-

emitters, alpha-emitters, and electron and neutron-capturing agents. In one
embodiment, gamma-emitters, positron-emitters, x-ray emitters, and
fluorescence-emitters are suitable for localization and/or therapy, while beta

and alpha-emitters and electron and neutron-capturing agents, such as boron
and uranium, also can be used for therapy.
[0288] The radionuclide used in the complex of the present invention may
be
suitable for therapeutic, diagnostic, or both therapeutic and diagnostic
purposes. Examples of appropriate metals include Ag, At, Au, Bi, Cu, Ga, Ho,
In, Lu, Pb, Pd, Pm, Pr, Rb, Re, Rh, Sc, Sr, Tc, Tl, Y, and Yb. Examples of the

radionuclide used for diagnostic purposes are Fe, Gd, 111In, 67Ga, or 68Ga.
In another embodiment, the radionuclide used for diagnostic purposes is
111In, or 67Ga. Examples of the radionuclide used for therapeutic purposes
are 166Ho, 165Dy, 90Y, 115mIn, 52Fe, or 72Ga. In one embodiment, the
radionuclide used for diagnostic purposes is 166Ho or 90Y. Examples of the

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
109
radionuclides used for both therapeutic and diagnostic purposes include
153Sm, 177Lu, 159Gd, 175Yb, or 47Sc. In one embodiment, the radionuclide
is 153Sm, 177Lu, 175Yb, or 159Gd.
[0289] Preferred metal radionuclides include 90Y, 99mTc, 111In, 47Sc,
67Ga,
51Cr, 177mSn, 67Cu, 167Tm, 97Ru, 188Re, 177Lu, 199Au, 47Sc, 67Ga,
51Cr, 177mSn, 67Cu, 167Tm, 95Ru, 188Re, 177Lu, 199Au, 203Pb and
141Ce.
[0290] In a particular embodiment, the metal ion of the antibody complex
is
selected from the group consisting of 90Y, 111In, 177Lu, 166Ho, 215Bi, and
225Ac.
[0291] Moreover, gamma-emitting radionuclides, such as 99mTc, 111In,
67Ga, and 169Yb have been approved or under investigation for diagnostic
imaging, while complexes of beta-emitters, such as 67Cu, 111Ag, 186Re, and
90Y are useful for the applications in tumor therapy. Also other useful
radionuclides include gamma-emitters, such as 99mTc, 111In, 67Ga, and
169Yb, and beta-emitters, such as 67Cu, 111Ag, 186Re, 188Re and 90Y, as
well as other radionuclides of interest such as 211At, 212Bi, 177Lu, 86Rb ,
105Rh, 153Sm, 198Au, 149Pm, 85Sr, 142Pr, 214Pb, 109Pd, 166Ho, 208T1,
and 44Sc.
[0292] In another embodiment, paramagnetic metal ions include ions of
transition and lanthanide metal, such as metals having atomic numbers of 21-
29, 42, 43, 44, or 57-71, in particular ions of Cr, V, Mn, Fe, Co, Ni, Cu, La,

Ce, Pr, Nd, Pm, Sm, Eu, Gd, Tb, Dy, Ho, Er, Tm, Yb, and Lu. The
paramagnetic metals used in the composition for magnetic resonance imaging
include the elements having atomic numbers of 22 to 29, 42, 44 and 58-70.
[0293] In another embodiment, fluorescent metal ions include lanthanides,
in
particular La, Ce, Pr, Nd, Pm, Sm, Eu, Gd, Tb, Dy, Ho, Er, Tm, Yb, and Lu.
[0294] In another embodiment, heavy metal-containing reporters may include
atoms of Mo, Bi, Si, and W.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
110
[0295] In a
preferred embodiment, an antibody complex of the present
invention comprises an antibody conjugate and a metal ion, wherein the
antibody of said conjugate is a human antibody.
[0296] In one embodiment, the chelator of the conjugate of the
invention is a
macrocyclic chelator, such as polyazamacrocyclic chelators, optionally
containing one or more carboxy, phosphonate, or phosphate groups. In
another embodiment, the chelator of the conjugate of the invention is a
chelator selected from the group consisting of DOTA, analogues of DOTA,
and derivatives of DOTA.
[0297] In one
embodiment, suitable chelator molecules include DOXA (1-
oxa-4,7,10-triazacyclododecanetriacetic acid), NOTA (1,4,7-
triazacyclononanetriacetic acid), TETA (1,4,8,11-

tetraazacyclotetradecanetetraacetic acid), and THT (4'-(3-amino-4-methoxy-
pheny1)-6,6"-bis(N',N1-dicarboxymethyl-N-methylhydra zino)-
2,2':6',2"-
terpyridine), and analogs and derivatives thereof. See, e.g., Ohmono et al.,
J.
Med. Chem. 35: 157-162 (1992); Kung et al., J. Nucl. Med. 25: 326-332
(1984); Jurisson et al., Chem. Rev. 93:1137-1156 (1993); and U.S. Patent No.
5,367,080. Other suitable chelators include chelating agents disclosed in U.S.

Patent Nos. 4,647,447; 4,687,659; 4,885,363; EP-A-71564; W089/00557; and
EP-A-232751.
[0298] In another embodiment, suitable macrocyclic carboxylic acid
chelators
which can be used in the present invention include 1,4,7,10-
tetraazacyclododecane-N,N',N",Nm-tetraacetic acid (DOTA); 1,4,8,12-
tetraazacyclopentadecane-N,N',N",Nm-tetraacetic acid (15N4);
triazacyclononane-N,N',N"-triacetic acid (9N3); 1,5,9-triazacyclododecane-
N,N',N"-triacetic acid (12N3); and 6-bromoacetamido-benzy1-1,4,8,11-
tetraazacyclotetadecane- N,N',N",N'"-tetraacetic acid (BAT).

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
111
Methods Of Preparing Antibody Complexes
[0299] An additional aspect of the present invention is directed to a
method of
preparing an antibody complex. In one embodiment, an antibody complex
may be prepared by reacting an antibody conjugate, as described herein, with a

metal ion, such as a radionuclide, which is able to associate noncovalently
with the conjugate. In another embodiment, the metal ion associates
noncovalently with the chelator moiety of said antibody conjugate. The
reaction between the antibody complex and the metal ion may occur in
solution, such as in a suitable buffered solution.
[0300] In one embodiment, a method for preparing an antibody complex
comprises reacting an antibody conjugate with a radionuclide. Such a method
comprises mixing, agitating, or preparing a solution comprising an antibody
conjugate and a radionuclide. In one embodiment, said solution further
comprises a buffer.
[0301] In another embodiment, a method for preparing an antibody complex
comprises reacting an antibody conjugate with a chelator complexed with a
metal ion. In this embodiment, a chelator is first complexed with a metal ion
according to known procedures in the art. For example, see U.S. Patent No.
5,654,361. The chelator can be an activated chelator as described above.
After the chelator is complexed with the metal ion and thereby forming a
chelator-metal ion complex, the antibody is reacted with the chelator-metal
ion
complex, using a procedure as described above for preparing an antibody
conjugate or using another procedure known in the art. According to the
method, the solution comprising the antibody and the chelator-metal ion
complex is mixed, agitated, or prepared, thereby forming said antibody
compex.
[0302] When preparing an antibody complex, the solution can be mixed,
agitated, or allowed to stand for a variable number of hours, depending upon a

number of variables, such factors including the identity of the chelator, the
identity of the antibody, the identity of the metal ion, the temperature of
the

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
112
reaction, the pH, the identity of the buffer, the molar ratio of the
reactants, the
purity of the available reagents, the presence of a catalyst, or activator,
and
other factors which would be evident to one of skill in the art. The solution
comprising an antibody conjugate and a metal ion can be mixed, agitated, or
allowed to stand for any amount of time that permits sufficient formation of
the antibody complex. In one embodiment of the above process, the solution
is mixed, agitated, or allowed to stand for about 0.5 minutes to about about
24
hours. In another embodiment, the solution is mixed, agitated, or allowed to
stand for about 1 hour to about 20 hours. In another embodiment, the solution
is mixed, agitated, or allowed to stand for about 2 hour to about 10 hours. In

another embodiment, the solution is mixed, agitated, or allowed to stand for
about 1 minute to about 1 hour. In another embodiment, the solution is mixed,
agitated, or allowed to stand for about 1 minute to about 30 minutes. In
another embodiment, the solution is mixed, agitated, or allowed to stand for
about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, or 55
minutes.
[0303] The pH of the solution used to prepare the antibody complex can
vary.
In one embodiment, the pH will be about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. In
another embodiment, said solution has a pH of about 4 to about 8. In another
embodiment, said solution has a pH of about 5 to about 7. In another
embodiment, said solution has a pH of about 5.5 to about 7. In another
embodiment, said solution has a pH of about 6.5.
[0304] The solution used to prepare the antibody complex may further
comprise a buffer. Buffers are well-known in the art. Suitable buffers for use

in the preparation of an antibody complex may include, but are not limited to,

citrate, acetate, phosphate, carbonate, diphosphate, glycyl-glycine-piperazine-

2HC1-Na0H; MES-Na0H-NaC1; TRIS-malic acid-NaOH; MES-NaOH;
ADA-Na0H-NaC1; ACES-Na0H-NaC1; ACES-NaOH-NaCl; BES-Na0H-
NaC1; MOPS-Na0H-NaC1; TES-NaOH-NaCl; MOPS-KOH; HEPES-Na0H-
NaC1; TRIS-HC1; HEPPSO-NaOH; BICINE-NaOH-NaCl; TAPS-Na0H-
NaC1; HEPPS (EPPS)-NaOH; TRICENE-NaOH; BICINE-NaOH; citric acid-
disodiumhydrogenphosphate; boric acid-citric acid-potassium dihydrogen

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
113
phosphate-Diethyl- barbituric acid-NaOH; citric acid-sodium citrate; sodium
acetate-acetic acid; potassium hydrogenphthalate-NaOH; cacodylic acid
sodium salt-HC1; potassium dihydrogen phosphate-disodium
hydrogenphosphate; potassium dihydrogen-phosphate-NaOH; sodium
dihydrogen phosphate- disodium hydrogen phosphate; imidazole-HC1; sodium
tetraborate-boric acid; 2-amino-2-
methy1-1,3-propanediol-HC1;
diethanolamine-HC1; potassium chloride-boric acid-NaOH; boric acid-Na0H-
KC1; glycine-NaOH; and sodium carbonate-sodium hydrogen carbonate.
[0305] The molar ratio of the antibody conjugate to the metal ion may
vary. In
one embodiment, the ratio of antibody conjugate to metal ion is from about
1:1000 to about 1000:1. In another embodiment, the ratio of antibody
conjugate to metal ion is from about 1:100 to about 100:1, in another
embodiment, from about 1:10 to about 10:1, in another embodiment, from
about 1:5 to about 5:1, in another embodiment about 1:2, in another
embodiment about 1:3, in another embodiment about 2:1, in another
embodiment about 1:1. In other embodiments, the ratio of antibody conjugate
to metal ion is about 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10.
[0306] Alternatively, the ratio of the antibody and the chelator-metal
ion
complex may vary. In one embodiment, the ratio of antibody to chelator-metal
ion complex is from about 1:1000 to 1000:1.
[0307] Radionuclides which can be used in the present invention are
described
and listed above and are well known and available commercially.
Additionally, several, known methods can be used to prepare the radionuclides
for use in the present invention. In one embodiment, the metal ion is in the
form of a salt, for example a chloride salt. Such salts are known in the art.
In
another embodiment, the metal ion salt is yttrium chloride or yttrium acetate.

In another embodiment, the metal ion used in the present invention
[0308] When preparing the complex of the present invention according to
one
embodiment, other metal ions which could compete for complexation with the
chelator are not present in significant amounts in the solution. For example,
when preparing an antibody complex comprising 90Y, Fe3+ is not present in

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
114
the solution in a significant amount or concentration. As used with reference
to the one or more competing metal ions in the present process, the term
significant amount refers to an amount or concentration which significantly
intereferes, retards, delays, inhibits, or prevents preparation of the
antibody
complex.
[0309] The method of preparing an antibody complex may further comprise a
step of removing excess metal ion from the reaction solution and/or the
antibody complex. In one embodiment, such a step comprises adding a
secondary chelating agent which is able to complex with excess metal ion in
the solution. Such a secondary chelating agent may include one or more
known chelating agents, for example, DTPA, EDTA, and Me0-DOTA-
glycine. In one embodiment, Me0-DOTA-glycine is used as a secondary
chelating agent.
[0310] The method of preparing an antibody complex may further comprise a
step of removing excess metal ion from the reaction solution comprising the
chelator-metal ion complex. In one embodiment, after the chelator-metal ion
complex is formed and is in solution, a secondary chelating agent which is
able to complex with excess metal ion is added. The solution is then eluted
through a DEAE-cellulose anion exhange resin (Sigma Chemical Co., St.
Louis, MO), which has been converted to acetate form to purify the neutral
species chelator-metal ion complex from the charged species, i.e., secondary
chelator-metal ion complex. The purified chelator-metal ion complex is then
used to prepared the antibody complex as described herein.
[0311] The secondary chelating agent can be added to the reaction mixture
as
a solid or as a solution. In one embodiment, the seconary chelating agent is
added in a buffered solution. In another embodiment of the present invention,
a buffer comprising an acetate buffer having a concentration of about 1 to
about 50 mM, preferably about 10 mM, having a NaC1 concentration of about
1 to about 500 mM, preferably about 140 mM, having a HSA concentration of
about 1% to about 20%, preferably about 7% to about 8%, more preferably
about 7.5%, having a pH of about 3-8, preferably about 6, and having a Me0-

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
115
DOTA-glycine concentration of about 0.01 mM to about 100 mM, preferably
about 1 mM, is added to the reaction solution after the antibody complex is
formed to a satisfactory level of completion. In one embodiment, the
secondary chelating agent is added about 5 minutes after the formation of the
antibody complex. In other embodiments, the secondary chelating agent is
added about 1, 2, 3, 4, 5, 6, 7 ,8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50 55,
or 60
minutes after formation of the antibody complex.
[0312] In another embodiment, the step of removing excess metal ion from
the
reaction solution and/or the antibody complex comprises subjecting the
reaction solution and/or the antibody complex to centrifugation. Subjecting
the reaction solution and/or the antibody complex to centrifugation can remove

non-chelated metal ions.
[0313] In another embodiment, the step of removing excess metal ion from
the
reaction solution and/or the antibody complex comprises washing the excess
metal ion from the reaction solution and/or the antibody complex with a
buffered solution. The washing may be repeated one or more times, as is
necessary. In one embodiment, the washing is repeated two or three times.
[0314] In one embodiment of the present method, two or more methods of
removing excess metal ion are used in combination to remove excess metal
ion from the step of removing excess metal ion from the reaction solution
and/or the antibody complex.
[0315] In another emodiment of the present method, for radiopharmaceutical
and radiotherapy applications, the antibody complex is prepared from a metal
in an oxidation state different from that of the desired complex. In this
case,
either a reducing agent or an oxidizing agent, depending on the oxidation
state
of the metal used and the oxidation state of the desired final product, is
added
to the reaction mixture to bring the metal to the desired oxidation state. The

oxidant or reductant can be used to form an intermediate complex in the
desired oxidation state but with labile ligands. These labile ligands can then

be displaced by the desired. chelating moiety of the present invention. In
another embodiment, the labile ligands are added to the reaction mixture along

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
116
with the reductant or oxidant and the desired ligand to achieve the change to
the desired oxidation state and chelation to the desired metal in a single
step.
[0316] In a further embodiment of the present invention, the method of
preparing an antibody complex comprises:
preparing a first solution comprising an antibody conjugate;
adding to said first solution a second solution comprising a metal ion
capable of complexing with said antibody conjugate;
mixing, agitating, or allowing to stand said first solution; and
optionally adding a secondary chelating agent, such as EDTA, to
complex with uncomplexed metal ion.
[0317] In an additional embodiment, the method of preparing an antibody
complex comprises:
combining a first solution, second solution, and a third solution,
wherein
said first solution comprises an antibody conjugate,
said second solution comprises an acetate buffer, and
said third solution comprises 90YC13;
mixing, agitating, or allowing to stand the combined solutions; and
adding to the combined solution a fourth solution, said fourth solution
comprising Me0-DOTA-NCS, human serum albumin (HSA), acetate
buffer, and NaCl.
Immunophenotyping
[0318] The antibodies such as the C35 antibodies of the invention may be
utilized for immunophenotyping of cell lines and biological samples. C35 is
useful as a tumor specific marker. Monoclonal antibodies directed against a
specific epitope, or combination of epitopes, will allow for the diagnosis of
cancer and screening of cellular populations. Various techniques can be
utilized using monoclonal antibodies to screen for cellular populations
expressing the marker(s), and include magnetic separation using antibody-

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
117
coated magnetic beads, "palming" with antibody attached to a solid matrix
(i.e., plate), and flow cytometry (See, e.g., U.S. Pat. No. 5,985,660; and
Morrison et al., Cell, 96:737-49 (1999)).
[0319] These techniques allow for the screening of particular populations
of
cells, such as might be found with hematological malignancies (e.g., C35-
associated cancers such as breast, ovarian, bladder, and colon cancers, and
melanoma).
Diagnosis And Imaging
[0320] Labeled C35 antibodies, and derivatives and analogs thereof, which
specifically bind to a polypeptide of interest can be used for diagnostic
purposes to detect, diagnose, or monitor diseases, disorders, and/or
conditions
associated with the aberrant expression and/or activity of a polypeptide of
the
invention (e.g., C35-associated cancers such as breast, ovarian, bladder, and
colon cancers, and melanoma). The invention provides for the detection of
aberrant expression of a polypeptide of interest, comprising (a) assaying the
expression of the polypeptide of interest in cells or body fluid of an
individual
using one or more antibodies specific to the polypeptide interest and (b)
comparing the level of gene expression with a standard gene expression level,
whereby an increase or decrease in the assayed polypeptide gene expression
level compared to the standard expression level is indicative of aberrant
expression.
[0321] The invention provides a diagnostic assay for diagnosing a
disorder,
comprising (a) assaying the expression of the polypeptide of interest in cells
or
body fluid of an individual using one or more antibodies specific to the
polypeptide interest and (b) comparing the level of gene expression with a
standard gene expression level, whereby an increase or decrease in the assayed

polypeptide gene expression level compared to the standard expression level is

indicative of a particular disorder. With respect to cancer, the presence of a

relatively high amount of transcript in biopsied tissue from an individual may

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
118
indicate a predisposition for the development of the disease, or may provide a

means for detecting the disease prior to the appearance of actual clinical
symptoms. A more definitive diagnosis of this type may allow health
professionals to employ preventative measures or aggressive treatment earlier
thereby preventing the development or further progression of the cancer.
[0322] Antibodies of the invention can be used to assay protein levels in
a
biological sample using classical immunohistological methods known to those
of skill in the art (e.g., see Jalkanen, et al., J. Cell. Biol. 101:976-985
(1985);
Jalkanen, et al., J. Cell Biol. 105:3087-3096 (1987)). Other antibody-based
methods useful for detecting protein gene expression include immunoassays,
such as the enzyme linked immunosorbent assay (ELISA) and the
radioimmunoassay (RIA). Suitable antibody assay labels are known in the art
and include enzyme labels, such as, glucose oxidase; radioisotopes, such as
iodine (1251, 1211), carbon (14C), sulfur (35S), tritium (3H), indium (1121n),

and technetium (99Tc); luminescent labels, such as luminol; and fluorescent
labels, such as fluorescein and rhodamine, and biotin.
[0323] One aspect of the invention is the detection and diagnosis of a
disease
or disorder associated with aberrant expression of a polypeptide of interest
in
an animal, preferably a mammal and most preferably a human. In one
embodiment, diagnosis comprises: a) administering (for example, parenterally,
subcutaneously, or intraperitoneally) to a subject an effective amount of a
labeled molecule which specifically binds to the polypeptide of interest; b)
waiting for a time interval following the administering for permitting the
labeled molecule to preferentially concentrate at sites in the subject where
the
polypeptide is expressed (and for unbound labeled molecule to be cleared to
background level); c) determining background level; and d) detecting the
labeled molecule in the subject, such that detection of labeled molecule above

the background level indicates that the subject has a particular disease or
disorder associated with aberrant expression of the polypeptide of interest.
Background level can be determined by various methods including, comparing

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
119
the amount of labeled molecule detected to a standard value previously
determined for a particular system.
[0324] It will be understood in the art that the size of the subject and
the
imaging system used will determine the quantity of imaging moiety needed to
produce diagnostic images. In the case of a radioisotope moiety, for a human
subject, the quantity of radioactivity injected will normally range from about
5
to 20 millicuries of 99mTc. The labeled antibody or antibody fragment will
then preferentially accumulate at the location of cells which contain the
specific protein. In vivo tumor imaging is described in S.W. Burchiel et al.,
"Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments."
(Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.
W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982).
[0325] Depending on several variables, including the type of label used
and
the mode of administration, the time interval following the administration for

permitting the labeled molecule to preferentially concentrate at sites in the
subject and for unbound labeled molecule to be cleared to background level is
6 to 48 hours or 6 to 24 hours or 6 to 12 hours. In another embodiment the
time interval following administration is 5 to 20 days or S to 10 days.
[0326] In an embodiment, monitoring of the disease or disorder is carried
out
by repeating the method for diagnosing the disease or disease, for example,
one month after initial diagnosis, six months after initial diagnosis, one
year
after initial diagnosis, etc.
[0327] Presence of the labeled molecule can be detected in the patient
using
methods known in the art for in vivo scanning. These methods depend upon
the type of label used. Skilled artisans will be able to determine the
appropriate
method for detecting a particular label. Methods and devices that may be used
in the diagnostic methods of the invention include, but are not limited to,
computed tomography (CT), whole body scan such as position emission
tomography (PET), magnetic resonance imaging (MRI), and sonography.
[0328] In a specific embodiment, the molecule is labeled with a
radioisotope
and is detected in the patient using a radiation responsive surgical
instrument

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
120
(Thurston et al., U.S. Pat. No. 5,441,050). In another embodiment, the
molecule is labeled with a fluorescent compound and is detected in the patient

using a fluorescence responsive scanning instrument. In another embodiment,
the molecule is labeled with a positron emitting metal and is detected in the
patent using positron emission-tomography. In yet another embodiment, the
molecule is labeled with a paramagnetic label and is detected in a patient
using
magnetic resonance imaging (MRI).
[0329] The antibodies of the present invention are useful for diagnosis,
treatment, prevention and/or prognosis of cancer disorders in mammals,
preferably humans. Such disorders include, but are not limited to, cancer,
neoplasms, tumors and/or as described under "Hyperproliferative Disorders"
below, especially C35-associated cancers such as breast, ovarian, bladder, and

colon cancers, and melanoma.
[0330] In particular, it is believed that certain tissues in mammals with
cancer
express significantly enhanced or reduced levels of normal or altered cancer
antigen expression and mRNA encoding the cancer associated polypeptide
when compared to a corresponding "standard" level. Further, it is believed
that
enhanced or depressed levels of the cancer associated polypeptide can be
detected in certain body fluids (e.g., sera, plasma, urine, and spinal fluid)
or
cells or tissue from mammals with such a cancer when compared to sera from
mammals of the same species not having the cancer.
[0331] For example, as disclosed herein, C35 expression is associated with
certain cancers (e.g., breast, ovarian, bladder, colon, and pancreatic
cancers,
and melanoma). Accordingly, antibodies (and antibody fragments) directed
against C35 may be used to quantitate or qualitate concentrations of cells
expressing C35. These antibodies additionally have diagnostic applications in
detecting abnormalities in the level of C35 expression, or abnormalities in
the
structure and/or temporal, tissue, cellular, or subcellular location of C35.
These diagnostic assays may be performed in vivo or in vitro, such as, for
example, on blood samples, biopsy tissue or autopsy tissue.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
121
[0332] Thus, the invention provides a diagnostic method useful during
diagnosis of a cancers, which involves measuring the expression level of the
cancer antigen polypeptide in tissue or other cells or body fluid from an
individual and comparing the measured expression level with a standard
cancer antigen expression level, whereby an increase in the expression level
compared to the standard is indicative of a disorder.
[0333] Where a diagnosis of a disorder, including diagnosis of a tumor,
has
already been made according to conventional methods, the present invention is
useful as a prognostic indicator, whereby patients exhibiting enhanced cancer
antigen gene expression will experience a worse clinical outcome relative to
patients expressing the gene at a level nearer the standard level.
[0334] By "assaying the expression level of the cancer associated
polypeptide"
is intended qualitatively or quantitatively measuring or estimating the level
of
the cancer antigen polypeptide in a first biological sample either directly
(e.g.,
by determining or estimating absolute protein level) or relatively (e.g., by
comparing to the cancer associated polypeptide level in a second biological
sample). Preferably, the cancer antigen polypeptide expression level in the
first
biological sample is measured or estimated and compared to a standard cancer
antigen polypeptide level, the standard being taken from a second biological
sample obtained from an individual not having the disorder or being
determined by averaging levels from a population of individuals not having the

disorder. As will be appreciated in the art, once a standard cancer antigen
polypeptide level is known, it can be used repeatedly as a standard for
comparison.
[0335] By "biological sample" is intended any biological sample obtained
from an individual, cell line, tissue culture, or other source containing
cancer
antigen polypeptides (including portions thereof). As indicated, biological
samples include body fluids (such as sera, plasma, urine, synovial fluid and
spinal fluid) which contain cells expressing cancer antigen polypeptides, and
other tissue sources found to express the full length or fragments thereof of
a

CA 02548180 2011-12-28
122
cancer antigen. Methods for obtaining tissue biopsies and body fluids from
mammals are well known in the art.
[0336] The present invention also relates to diagnostic assays such as
quantitative and diagnostic assays for detecting levels of cancer antigen
polypeptides, in a biological sample (e.g., cells and tissues), including
determination of normal and abnormal levels of polypeptides. Thus, for
instance, a diagnostic assay in accordance with the invention for detecting
over-expression of cancer antigens compared to normal control tissue samples
may be used to detect the presence of tumors. Assay techniques that can be
used to determine levels of a polypeptide, such as a cancer antigen
polypeptide
of the present invention in a sample derived from a host are well-known to
those of skill in the art. Such assay methods include radioimmunoassays,
competitive-binding assays, Western Blot analysis and ELISA assays.
Assaying cancer antigen polypeptide levels in a biological sample can occur
using any art-known method.
[0337] Cancer antigen polypeptide expression in tissues can also be studied
with classical immunohistological methods (Jalkanen et al., J. Cell. Biol.
101:976-985 (1985); Jalkanen, M., et al., J. Cell . Biol. 105:3087-3096
(1987)). Other antibody-based methods useful for detecting cancer antigen
polypeptide gene expression include immunoassays, such as the enzyme linked
immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable
antibody assay labels are known in the art and include enzyme labels, such as,

glucose oxidase, and radioisotopes, such as iodine (1251, 1211), carbon (14C),

sulfur (35S), tritium (3H), indium (1121n), and technetium (99mTc), and
fluorescent labels, such as fluorescein and rhodamine, and biotin.
[0338] The tissue or cell type to be analyzed will generally include those
which are known, or suspected, to express the cancer antigen gene. The protein

isolation methods employed herein may, for example, be such as those
described in Harlow and Lane (Harlow, E. and Lane, D., 1988, "Antibodies: A
Laboratory Manual", Cold Spring Harbor Laboratory Press, Cold Spring
Harbor, New York).

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
123
The isolated cells can be derived from cell culture or from a patient. The
analysis of cells taken from culture may be a necessary step in the assessment

of cells that could be used as part of a cell-based gene therapy technique or,

alternatively, to test the effect of compounds on the expression of the cancer

antigen gene.
[0339] In an additional preferred embodiment, antibodies, or fragments of
antibodies directed to a conformational epitope of a cancer antigen may be
used to quantitatively or qualitatively detect the presence of cancer antigen
gene products or conserved variants or peptide fragments thereof. This can be
accomplished, for example, by immunofluorescence techniques employing a
fluorescently labeled antibody coupled with light microscopic, flow
cytometric, or fluorimetric detection.
[0340] The antibodies (or fragments thereof), and/or cancer antigen
polypeptides of the present invention may, additionally, be employed
histologically, as in immunofluorescence, immunoelectron microscopy or non-
immunological assays, for in situ detection of cancer antigen gene products or

conserved variants or peptide fragments thereof. In situ detection may be
accomplished by removing a histological specimen from a patient, and
applying thereto a labeled antibody or cancer antigen polypeptide of the
present invention. The antibody (or fragment thereof) or cancer antigen
polypeptide is preferably applied by overlaying the labeled antibody (or
fragment) onto a biological sample. Through the use of such a procedure, it is

possible to determine not only the presence of the cancer antigen gene
product,
or conserved variants or peptide fragments, or cancer antigen polypeptide
binding, but also its distribution in the examined tissue. Using the present
invention, those of ordinary skill will readily perceive that any of a wide
variety of histological methods (such as staining procedures) can be modified
in order to achieve such in situ detection.
[0341] Immunoassays and non-immunoassays for cancer antigen gene
products or conserved variants or peptide fragments thereof will typically
comprise incubating a sample, such as a biological fluid, a tissue extract,

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
124
freshly harvested cells, or lysates of cells which have been incubated in cell

culture, in the presence of a detectably labeled antibody capable of binding
cancer antigen gene products or conserved variants or peptide fragments
thereof, and detecting the bound antibody by any of a number of techniques
well-known in the art.
[0342] The biological sample may be brought in contact with and
immobilized
onto a solid phase support or carrier such as nitrocellulose, or other solid
support which is capable of immobilizing cells, cell particles or soluble
proteins. The support may then be washed with suitable buffers followed by
treatment with the detectably labeled anti-cancer antigen antibody or
detectable cancer antigen polypeptide. The solid phase support may then be
washed with the buffer a second time to remove unbound antibody or
polypeptide. Optionally the antibody is subsequently labeled. The amount of
bound label on solid support may then be detected by conventional means.
[0343] By "solid phase support or carrier" is intended any support capable
of
binding an antigen or an antibody. Well-known supports or carriers include
glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases,
natural and modified celluloses, polyacrylamides, gabbros, and magnetite. The
nature of the carrier can be either soluble to some extent or insoluble for
the
purposes of the present invention. The support material may have virtually any

possible structural configuration so long as the coupled molecule is capable
of
binding to an antigen or antibody. Thus, the support configuration may be
spherical, as in a bead, or cylindrical, as in the inside surface of a test
tube, or
the external surface of a rod. Alternatively, the surface may be flat such as
a
sheet, test strip, etc. Preferred supports include polystyrene beads. Those
skilled in the art will know many other suitable carriers for binding antibody
or
antigen, or will be able to ascertain the same by use of routine
experimentation.
[0344] The binding activity of a given lot of anti-cancer antigen antibody
or
cancer antigen polypeptide may be determined according to well known
methods. Those skilled in the art will be able to determine operative and

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
125
optimal assay conditions for each determination by employing routine
experimentation.
[0345] In addition to assaying cancer antigen polypeptide levels or
polynucleotide levels in a biological sample obtained from an individual,
cancer antigen polypeptide or polynucleotide can also be detected in vivo by
imaging. For example, in one embodiment of the invention, cancer antigen
polypeptide and/or anti-cancer antigen antibodies are used to image diseased
cells, such as neoplasms. In another embodiment, cancer antigen
polynucleotides of the invention (e.g., polynucleotides complementary to all
or
a portion of cancer antigen mRNA) and/or anti-cancer antigen antibodies (e.g.,

antibodies directed to any one or a combination of the epitopes of cancer
antigens, antibodies directed to a conformational epitope of cancer antigens,
antibodies directed to the full length polypeptide expressed on the cell
surface
of a mammalian cell) are used to image diseased or neoplastic cells.
[0346] Antibody labels or markers for in vivo imaging of cancer antigen
polypeptides include those detectable by X-radiography, NMR, MRI, CAT-
scans or ESR. For X-radiography, suitable labels include radioisotopes such as

barium or cesium, which emit detectable radiation but are not overtly harmful
to the subject. Suitable markers for NMR and ESR include those with a
detectable characteristic spin, such as deuterium, which may be incorporated
into the antibody by labeling of nutrients for the relevant hybridoma. Where
in
vivo imaging is used to detect enhanced levels of cancer antigen polypeptides
for diagnosis in humans, it may be preferable to use human antibodies or
"humanized" chimeric monoclonal antibodies. Such antibodies can be
produced using techniques described herein or otherwise known in the art. For
example methods for producing chimeric antibodies are known in the art. See,
for review, Morrison, Science 229:1202 (1985); Oi et al., BioTechniques
4:214 (1986); Cabilly et al., U.S. Pat. No. 4,816,567; Taniguchi et al., EP
171496; Morrison et al., EP 173494; Neuberger et al., WO 8601533; Robinson
et al., WO 8702671; Boulianne et al., Nature 312:643 (1984); Neuberger et al.,

Nature 314:268 (1985).

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
126
[0347] Additionally, any cancer antigen polypeptides whose presence can
be
detected, can be administered. For example, cancer antigen polypeptides
labeled with a radio-opaque or other appropriate compound can be
administered and visualized in vivo, as discussed, above for labeled
antibodies. Further such cancer antigen polypeptides can be utilized for in
vitro diagnostic procedures.
[0348] A cancer antigen polypeptide-specific antibody or antibody
fragment
which has been labeled with an appropriate detectable imaging moiety, such as
a radioisotope (for example, 1311, 112In, 99mTc), a radio-opaque substance,
or a material detectable by nuclear magnetic resonance, is introduced (for
example, parenterally, subcutaneously or intraperitoneally) into the mammal to

be examined for a disorder. It will be understood in the art that the size of
the
subject and the imaging system used will determine the quantity of imaging
moiety needed to produce diagnostic images. In the case of a radioisotope
moiety, for a human subject, the quantity of radioactivity injected will
normally range from about 5 to 20 millicuries of 99mTc. The labeled antibody
or antibody fragment will then preferentially accumulate at the location of
cells
which contain cancer antigen protein. In vivo tumor imaging is described in S.

W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled Antibodies and
Their Fragments" (Chapter 13 in Tumor Imaging: The Radiochemical
Detection of Cancer, S. W. Burchiel and B. A. Rhodes, eds., Masson
Publishing Inc. (1982)).
[0349] With respect to antibodies, one of the ways in which the anti-
cancer
antigen antibody can be detectably labeled is by linking the same to an enzyme

and using the linked product in an enzyme immunoassay (ETA) (Voller, A.,
"The Enzyme Linked Immunosorbent Assay (ELISA)", 1978, Diagnostic
Horizons 2:1-7, Microbiological Associates Quarterly Publication,
Walkersville, Md.); Voller et al., J. Clin. Pathol. 31:507-520 (1978); Butler,
J.
E., Meth. Enrymol. 73:482-523 (1981); Maggio, E. (ed.), 1980, Enzyme
Immunoassay, CRC Press, Boca Raton, Fla.,; Ishikawa, E. et al., (eds.), 1981,
Enzyme Immunoassay, Kgaku Shoin, Tokyo). The enzyme, which is bound to

CA 02548180 2011-12-28
127
the antibody will react with an appropriate substrate, preferably a
chromogenic
substrate, in such a manner as to produce a chemical moiety which can be
detected, for example, by spectrophotometric, fluorimetric or by visual means.

Enzymes which can be used to detectably label the antibody include, but are
not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid

isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate,
dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline
phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease,
urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and
acetylcholinesterase. Additionally, the detection can be accomplished by
colorimetric methods which employ a cliromogenic substrate for the enzyme.
Detection may also be accomplished by visual comparison of the extent of
enzymatic reaction of a substrate in comparison with similarly prepared
standards.
[0350] Detection may also be accomplished using any of a variety of other
immunoassays. For example, by radioactively labeling the antibodies or
antibody fragments, it is possible to detect cancer antigens through the use
of a
radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of
Radioimmunoassays, Seventh Training Course on Radioligand Assay
Techniques, The Endocrine Society, March, 1986).
The radioactive isotope can be detected by means including,
but not limited to, a gamma counter, a scintillation counter, or
autoradiogaphy.
[0351] It is also possible to label the antibody with a fluorescent
compound.
When the fluorescently labeled antibody is exposed to light of the proper wave

length, its presence can then be detected due to fluorescence. Among the most
commonly used fluorescent labeling compounds are fluorescein
isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin,
ophthaldehyde and fluorescamine.
[0352] The antibody can also be detectably labeled using fluorescence
emitting metals such as 152Eu, or others of the lanthanide series. These
metals

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
128
can be attached to the antibody using such metal chelating groups as
diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid
(EDTA).
[0353] The antibody also can be detectably labeled by coupling it to a
chemiluminescent compound. The presence of the chemiluminescent-tagged
antibody is then determined by detecting the presence of luminescence that
arises during the course of a chemical reaction. Examples of particularly
useful
chemiluminescent labeling compounds are luminol, isoluminol, theromatic
acridinium ester, imidazole, acridinium salt and oxalate ester.
[0354] Likewise, a bioluminescent compound may be used to label the
antibody of the present invention. Bioluminescence is a type of
chemiluminescence found in biological systems in, which a catalytic protein
increases the efficiency of the chemiluminescent reaction. The presence of a
bioluminescent protein is determined by detecting the presence of
luminescence. Important bioluminescent compounds for purposes of labeling
are luciferin, luciferase and aequorin.
[0355] In general, cancer may be detected in a patient based on the
presence of
one or more cancer antigen proteins of the invention and/or polynucleotides
encoding such proteins in a biological sample (for example, blood, sera,
urine,
and/or tumor biopsies) obtained from the patient. In other words, such
proteins
and/or polynucleotides may be used as markers to indicate the presence or
absence of cancer. Cancers that may be diagnosed, and/or prognosed using the
compositions of the invention include but are not limited to, colorectal
cancer,
breast cancer, ovarian cancer, prostate cancer, pancreatic cancer, lung
cancer,
liver cancer, uterine cancer, and/or skin cancer. The binding agents provided
herein generally permit detection of the level of antigen that binds to the
agent
in the biological sample. Polynucleotide primers and probes may be used to
detect the level of MRNA encoding cancer antigen polypeptides, which is also
indicative of the presence or absence of cancer. In general, cancer antigen
polypeptides should be present at a level that is at least three fold higher
in
diseased tissue than in normal tissue.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
129
[0356] There are a variety of assay formats known to those of ordinary
skill in
the art for using a binding agent to detect polypeptide markers in a sample.
See, e.g., Harlow and Lane, supra. In general, the presence or absence of a
disease in a patient may be determined by (a) contacting a biological sample
obtained from a patient with a binding agent; (b) detecting in the sample a
level of polypeptide that binds to the binding agent; and (c) comparing the
level of polypeptide with a predetermined cut-off value.
[0357] In a preferred embodiment, the assay involves the use of binding
agent
immobilized on a solid support to bind to and remove the cancer antigen
polypeptide of the invention from the remainder of the sample. The bound
polypeptide may then be detected using a detection reagent that contains a
reporter group and specifically binds to the binding agent/polypeptide
complex. Such detection reagents may comprise, for example, a binding agent
that specifically binds to the polypeptide or an antibody or other agent that
specifically binds to the binding agent, such as an anti-immunoglobulin,
protein G, protein A or a lectin. Alternatively, a competitive assay may be
utilized, in which a polypeptide is labeled with a reporter group and allowed
to
bind to the immobilized binding agent after incubation of the binding agent
with the sample. The extent to which components of the sample inhibit the
binding of the labeled polypeptide to the binding agent is indicative of the
reactivity of the sample with the immobilized binding agent. Suitable
polypeptides for use within such assays include cancer antigen polypeptides
and portions thereof, or antibodies, to which the binding agent binds, as
described above.
[0358] The solid support may be any material known to those of skill in
the art
to which cancer antigen polypeptides of the invention may be attached. For
example, the solid support may be a test well in a microtiter plate or a
nitrocellulose or other suitable membrane. Alternatively, the support may be a

bead or disc, such as glass fiberglass, latex or a plastic material such as
polystyrene or polyvinylchloride. The support may also be a magnetic particle
or a fiber optic sensor, such as those disclosed, for example, in U.S. Pat.
No.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
130
5,359,681. The binding agent may be immobilized on the solid support using a
variety of techniques known to those of skill in the art, which are amply
described in the patent and scientific literature. In the context of the
present
invention, the term "immobilization" refers to both noncovalent association,
such as adsorption, and covalent attachment (which may be a direct linkage
between the agent and functional groups on the support or may be a linkage by
way of a cross-linking agent). Immobilization by adsorption to a well in a
microtiter plate or to a membrane is preferred. In such cases, adsorption may
be achieved by contacting the binding agent, in a suitable buffer, with the
solid
support for the suitable amount of time. The contact time varies with
temperature, but is typically between about 1 hour and about 1 day. In
general,
contacting a well of plastic microtiter plate (such as polystyrene or
polyvinylchloride) with an amount of binding agent ranging from about 10 ng
to about 10 ug, and preferably about 100 ng to about 1 ug, is sufficient to
immobilize an adequate amount of binding agent.
[0359] Covalent attachment of binding agent to a solid support may
generally
be achieved by first reacting the support with a bifunctional reagent that
will
react with both the support and a functional group, such as a hydroxyl or
amino group, on the binding agent. For example, the binding agent may be
covalently attached to supports having an appropriate polymer coating using
benzoquinone or by condensation of an aldehyde group on the support with an
amine and an active hydrogen on the binding partner (see, e.g., Pierce
Immunotechnology Catalog and Handbook, 1991, at Al2-A13).
Therapeutic/Prophylactic Administration And Composition
[0360] In one embodiment, the entire antibody dose is provided in a
single
bolus. Alternatively, the dose can be provided by multiple administrations,
such as an extended infusion method or by repeated injections administered
over a span of hours or days, for example, a span of about 2 to about 4 days.
Also see Examples 5, 9 and 10, and Tables 6-9. =

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
131
[0361] Formulations and methods of administration that can be employed
when the compound comprises a nucleic acid or an immunoglobulin are
described above; additional appropriate formulations and routes of
administration can be selected from among those described herein below.
[0362] Various delivery systems are known and can be used to administer a
compound of the invention, e.g., encapsulation in liposomes, microparticles,
microcapsules, recombinant cells capable of expressing the compound,
receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem.
262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral
or
other vector, etc. Methods of introduction include but are not limited to
intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous,
intranasal, epidural, and oral routes. The compounds or compositions may be
administered by any convenient route, for example by infusion or bolus
injection, by absorption through epithelial or mucocutaneous linings (e.g.,
oral
mucosa, rectal and intestinal mucosa, etc.) and may be administered together
with other biologically active agents. Administration can be systemic or
local.
In addition, it may be desirable to introduce the pharmaceutical compounds or
compositions of the invention into the central nervous system by any suitable
route, including intraventricular and intrathecal injection; intraventricular
injection may be facilitated by an intraventricular catheter, for example,
attached to a reservoir, such as an Ommaya reservoir. Pulmonary
administration can also be employed, e.g., by use of an inhaler or nebulizer,
and formulation with an aerosolizing agent.
[0363] In a specific embodiment, it may be desirable to administer the
pharmaceutical compounds or compositions of the invention locally to the area
in need of treatment; this may be achieved by, for example, and not by way of
limitation, local infusion during surgery, topical application, e.g., in
conjunction with a wound dressing after surgery, by injection, by means of a
catheter, by means of a suppository, or by means of an implant, said implant
being of a porous, non-porous, or gelatinous material, including membranes,
such as sialastic membranes, or fibers. Preferably, when administering a

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
132
protein, including an antibody, of the invention, care must be taken to use
materials to which the protein does not absorb.
[0364] In another embodiment, the compound or composition can be delivered
in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533
(1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and
Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365
(1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
[0365] In yet another embodiment, the compound or composition can be
delivered in a controlled release system. In one embodiment, a pump may be
used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987);
Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med.
321:574 (1989)). In another embodiment, polymeric materials can be used (see
Medical Applications of Controlled Release, Langer and Wise (eds.), CRC
Pres., Boca Raton, Florida (1974); Controlled Drug Bioavailability, Drug
Product Design and Performance, Smolen and Ball (eds.), Wiley, New York
(1984); Ranger and Peppas, J., Macromol. Sci. Rev. Macromol. Chem. 23:61
(1983); see also Levy et al., Science 228:190 (1985); During et al., Arm.
Neurol. 25:351 (1989); Howard et al., J.Neurosurg. 71:105 (1989)). In yet
another embodiment, a controlled release system can be placed in proximity of
the therapeutic target, i.e., the brain, thus requiring only a fraction of the

systemic dose (see, e.g., Goodson, in Medical Applications of Controlled
Release, supra, vol. 2, pp. 115-138 (1984)).
[0366] Other controlled release systems are discussed in the review by
Langer
(Science 249:1527-1533 (1990)).
[0367] In a specific embodiment where the compound of the invention is a
nucleic acid encoding a protein, the nucleic acid can be administered in vivo
to
promote expression of its encoded protein, by constructing it as part of an
appropriate nucleic acid expression vector and administering it so that it
becomes intracellular, e.g., by use of a retroviral vector (see U.S. Pat. No.
4,980,286), or by direct injection, or by use of microparticle bombardment
(e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
133
receptors or transfecting agents, or by administering it in linkage to a
homeobox- like peptide which is known to enter the nucleus (see e.g., Joliot
et
al., Proc. Natl. Acad. Sci. USA 88:1864-1868 (1991)), etc. Alternatively, a
nucleic acid can be introduced intracellularly and incorporated within host
cell
DNA for expression, by homologous recombination.
[0368] The present invention also provides pharmaceutical compositions.
Such compositions comprise a therapeutically effective amount of a
compound, and a pharmaceutically acceptable carrier. In a specific
embodiment, the term "pharmaceutically acceptable" means approved by a
regulatory agency of the Federal or a state government or listed in the U.S.
Pharmacopeia or other generally recognized pharmacopeia for use in animals,
and more particularly in humans. The term "carrier" refers to a diluent,
adjuvant, excipient, or vehicle with which the therapeutic is administered.
Such pharmaceutical carriers can be sterile liquids, such as water and oils,
including those of petroleum, animal, vegetable or synthetic origin, such as
peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a
preferred carrier when the pharmaceutical composition is administered
intravenously. Saline solutions and aqueous dextrose and glycerol solutions
can also be employed as liquid carriers, particularly for injectable
solutions.
Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose,
gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol
monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene,
glycol, water, ethanol and the like. The composition, if desired, can also
contain minor amounts of wetting or emulsifying agents, or pH buffering
agents. These compositions can take the form of solutions, suspensions,
emulsion, tablets, pills, capsules, powders, sustained-release formulations
and
the like. The composition can be formulated as a suppository, with traditional

binders and carriers such as triglycerides. Oral formulation can include
standard carriers such as pharmaceutical grades of mannitol, lactose, starch,
magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc.
Examples of suitable pharmaceutical carriers are described in "Remington's

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
134
Pharmaceutical Sciences" by E. W. Martin. Such compositions will contain a
therapeutically effective amount of the compound, preferably in purified form,

together with a suitable amount of carrier so as to provide the form for
proper
administration to the patient. The formulation should suit the mode of
administration.
[0369] In a preferred embodiment, the composition is formulated in
accordance with routine procedures as a pharmaceutical composition adapted
for intravenous administration to human beings. Typically, compositions for
intravenous administration are solutions in sterile isotonic aqueous buffer.
Where necessary, the composition may also include a solubilizing agent and a
local anesthetic such as lignocaine to ease pain at the site, of the
injection.
Generally, the ingredients are supplied either separately or mixed together in

unit dosage form, for example, as a dry lyophilized powder or water free
concentrate in a hermetically sealed container such as an ampoule or sachette
indicating the quantity of active agent. Where the composition is to be
administered by infusion, it can be dispensed with an infusion bottle
containing sterile pharmaceutical grade water or saline. Where the
composition is administered by injection, an ampoule of sterile water for
injection or saline can be provided so that the ingredients may be mixed prior

to administration.
[0370] The compounds of the invention can be formulated as neutral or
salt
forms. Pharmaceutically acceptable salts include those formed with anions
such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric
acids, etc., and those formed with cations such as those derived from sodium,
potassium, ammonium, calcium, ferric hydroxides, isopropylamine,
triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
[0371] The amount of the compound of the invention which will be
effective
in the treatment, inhibition and prevention of a disease or disorder
associated
with aberrant expression and/or activity of a polypeptide of the invention can

be determined by standard clinical techniques. In addition, in vitro assays
may
optionally be employed to help identify optimal dosage ranges. The precise

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
135
dose to be employed in the formulation will also depend on the route of
administration, and the seriousness of the disease or disorder, and should be
decided according to the judgment of the practitioner and each patient's
circumstances. Effective doses may be extrapolated from dose-response curves
derived from in vitro or animal model test systems.
[0372] For antibodies, the dosage administered to a patient is typically
0.1
mg/kg to 100 mg/kg of the patient's body weight. Preferably, the dosage
administered to a patient is between 0.1 mg/kg and 20 mg/kg of the patient's
body weight, more preferably 1 mg/kg to 10 mg/kg of the patient's body
weight. Generally, human antibodies have a longer half-life within the human
body than antibodies from other species due to the immune response to the
foreign polypeptides. Thus, lower dosages of human antibodies and less
frequent administration is often possible. Further, the dosage and frequency
of
administration of antibodies of the invention may be reduced by enhancing
uptake and tissue penetration (e.g., into the brain) of the antibodies by
modifications such as, for example, lipidation. Also see Example 5.
[0373] The invention also provides a pharmaceutical pack or kit comprising
one or more containers filled with one or more of the ingredients of the
pharmaceutical compositions of the invention. Optionally associated with such
container(s) can be a notice in the form prescribed by a governmental agency
regulating the manufacture, use or sale of pharmaceuticals or biological
products, which notice reflects approval by the agency of manufacture, use or
sale for human administration.
[0374] Antibodies can be used to assay levels of polypeptides encoded by
polynucleotides of the invention in a biological sample using classical
immunohistological methods known to those of skill in the art (e.g., see
Jalkanen, et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, et al., J.
Cell.
Biol. 105:3087-3096 (1987)). Other antibody-based methods useful for
detecting protein gene expression include immunoassays, such as the enzyme
linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
Suitable antibody assay labels are known in the art and include enzyme labels,

CA 02 5 4 81 8 0 2 0 0 6-0 6-01
WO 2005/055936
PCT/US2004/040573
136
such as, glucose oxidase; radioisotopes, such as iodine (1311, 1251, 1231,
121D,
carbon (14C), sulfur (35S), tritium (3H), indium (115m1n, 113in, 112 11114
and
technetium (99Tc, 99mTc), thallium col r) gallium (68Ga, 67Ga), palladium
(103P
d), molybdenum (99Mo), xenon (133Xe), fluorine (8F), 153SM, 1771,11,
159Gd, 149PM, 14012, 175yb, 166H0, 90-y, 47se, 186Re, 88Re, 142pr, 105- ,
97Ru;
luminescent labels, such as luminol; and fluorescent labels, such as
fluorescein
and rhodamine, and biotin.
[0375] In addition to assaying levels of polypeptide of the present
invention in
a biological sample, proteins can also be detected in vivo by imaging.
Antibody labels or markers for in vivo imaging of protein include those
detectable by X-radiography, NMR or ESR. For X-radiography, suitable labels
include radioisotopes such as barium or cesium, which emit detectable
radiation but are not overtly harmful to the subject. Suitable markers for NMR

and ESR include those with a detectable characteristic spin, such as
deuterium,
which may be incorporated into the antibody by labeling of nutrients for the
relevant hybridoma.
[0376] A protein-specific antibody or antibody fragment which has been
labeled with an appropriate detectable imaging moiety, such as a radioisotope
(for example, 1311, 112In, 99mTc, (1311, 1251, 1231, )
121".1.-',
carbon (14C), sulfur (35S),
tritium (3H), indium (115mIn, 13m1n, 112m, 1111n),
and technetium (99Tc,
99mTc), thallium (201Ti), gallium (68Ga, 67Ga), palladium (103Pd), molybdenum
(99Mo), xenon (133Xe), fluorine (18F, 1535114 177LU, 59Gd, 149pm, 140La,
1751b,
166H0, 90y, 47se, 186Re, 188Re, 142pr, 105-," , 9
-7Ru), a radio-opaque substance, or
a material detectable by nuclear magnetic resonance, is introduced (for
example, parenterally, subcutaneously or intraperitoneally) into the mammal to

be examined for immune system disorder. It will be understood in the art that
the size of the subject and the imaging system used will determine the
quantity
of imaging moiety needed to produce diagnostic images. In the case of a
radioisotope moiety, for a human subject, the quantity of radioactivity
injected
will normally range from about 5 to 20 millicuries of 99mTc. The labeled
antibody or antibody fragment will then preferentially accumulate at the

CA 02548180 2011-12-28
137
location of cells which express the polypeptide encoded by a polynucleotide of

the invention. In vivo tumor imaging is described in S.W. Burchiel et al.,
"Immunopharmacokinetics of Radiolabeled Antibodies and Their agments"
(Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.
W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)).
[0377] In one embodiment, the invention provides a method for the specific
delivery of compositions of the invention to cells by administering
polypeptides of the invention (e.g., polypeptides encoded by polynucleotides
of the invention and/or antibodies) that are associated with heterologous
polypeptides or nucleic acids. In one example, the invention provides a method

for delivering a therapeutic protein into the targeted cell. In another
example,
the invention provides a method for delivering a single stranded nucleic acid
(e.g., antisense or ribozymes) or double stranded nucleic acid (e.g., DNA that

can integrate into the cell's genome or replicate episomally and that can be
transcribed) into the targeted cell. =
[0378] Techniques known in the art may be applied to label polypeptides of
the invention (including antibodies). Such techniques include, but are not
limited to, the use of bifunctional conjugating agents (see e.g., U.S. Pat.
Nos.
5,756,065; 5,714,631; 5,696,239; 5,652,361; 5,505,931; 5,489,425; 5,435,990;
5,428,139; 5,342,604; 5,274,119; 4,994,560; and 5,808,003).
Gene Therapy
[0379] In a specific embodiment, nucleic acids comprising sequences
encoding antibodies such as C35 antibodies, or functional derivatives thereof,

are administered to treat, inhibit or prevent a disease or disorder associated

with aberrant expression and/or activity of C35, by way of gene therapy. Gene
therapy refers to therapy performed by the administration to a subject of an
expressed or expressible nucleic acid. In this embodiment of the invention,
the
nucleic acids produce their encoded protein that mediates a therapeutic
effect.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
138
[0380] Any of the methods for gene therapy available in the art can be
used
according to the present invention. Exemplary methods are described below.
[0381] For general reviews of the methods of gene therapy, see Goldspiel
et
al., Clinical Pharmacy 12:488-505 (1993); Wu and Wu, Biotherapy 3:87-95
(1991); Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993);
Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann. Rev.
Biochem. 62:191-217 (1993); May, TIBTECH 11(5):155-215 (1993). Methods
commonly known in the art of recombinant DNA technology which can be
used are described in Ausubel et al. (eds.), Current Protocols in Molecular
Biology, John Wiley & Sons, NY (1993); and Kriegler, Gene Transfer and
Expression, A Laboratory Manual, Stockton Press, NY (1990).
[0382] In a preferred aspect, the compound comprises nucleic acid
sequences
encoding an antibody, said nucleic acid sequences being part of expression
vectors that express the antibody or fragments or chimeric proteins or heavy
or
light chains thereof in a suitable host. In particular, such nucleic acid
sequences have promoters operably linked to the antibody coding region, said
promoter being inducible or constitutive, and, optionally, tissue-specific. In

another particular embodiment, nucleic acid molecules are used in which the
antibody coding sequences and any other desired sequences are flanked by
regions that promote homologous recombination at a desired site in the
genome, thus providing for intrachromosomal expression of the antibody
encoding nucleic acids (Koller and Smithies, Proc. Natl. Acad. Sci. USA
86:8932-8935 (1989); Zijlstra et al., Nature 342:435-438 (1989). In specific
embodiments, the expressed antibody molecule is a single chain antibody;
alternatively, the nucleic acid' sequences include sequences encoding both the

heavy and light chains, or fragments thereof, of the antibody.
[0383] Delivery of the nucleic acids into a patient may be either direct,
in
which case the patient is directly exposed to the nucleic acid or nucleic acid-

carrying vectors, or indirect, in which case, cells are first transformed with
the
nucleic acids in vitro, then transplanted into the patient. These two
approaches
are known, respectively, as in vivo or ex vivo gene therapy.

CA 02548180 2011-12-28
139
[0384] In a specific embodiment, the nucleic acid sequences are directly
administered in vivo, where it is expressed to produce the encoded product.
This can be accomplished by any of numerous methods known in the art, e.g.,
by constructing them as part of an appropriate nucleic acid expression vector
and administering it so that they become intracellular, e.g., by infection
using
defective or attenuated retrovirals or other viral vectors (see U.S. Pat. No.
4,980,286), or by direct injection of naked DNA, or by use of microparticle
TM TM
bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or
cell-surface receptors or transfecting agents, encapsulation in liposomes,
microparticles, or microcapsules, or by administering them in linkage to a
peptide which is known to enter the nucleus, by administering it in linkage to
a
ligand subject to receptor-mediated endocytosis (see, e.g., Wu and Wu, J.
Biol.
Chem. 262:4429-4432 (1987)) (which can be used to target cell types
specifically expressing the receptors), etc. In another embodiment, nucleic
acid-ligand complexes can be formed in which the ligand comprises a
fusogenic viral peptide to disrupt endosomes, allowing the nucleic acid to
avoid lysosomal degradation. In yet another embodiment, the nucleic acid can
be targeted in vivo for cell specific uptake and expression, by targeting a
specific receptor (see, e.g., PCT Publications WO 92/06180; WO 92/22635;
W092/20316; W093/14188, WO 93/20221). Alternatively, the nucleic acid
can be introduced intracellularly and incorporated within host cell DNA for
expression, by homologous recombination (Koller and Smithies, Proc. Natl.
Acad. Sci. USA 86:8932-8935 (1989); Zijlstra et al., Nature 342:435-438
(1989)).
[0385] In a specific embodiment, viral vectors that contains nucleic acid
sequences encoding an antibody of the invention are used. For example, a
retroviral vector can be used (see Miller et al., Meth. Enzymol. 217:581-599
(1993)). These retroviral vectors contain the components necessary for the
correct packaging of the viral genome and integration into the host cell DNA.
The nucleic acid sequences encoding the antibody to be used in gene therapy
are cloned into one or more vectors, which facilitates delivery of the gene
into

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
140
a patient. More detail about retroviral vectors can be found in Boesen et al.,

Biotherapy 6:291-302 (1994), which describes the use of a retroviral vector to

deliver the mdrl gene to hematopoietic stem cells in order to make the stem
cells more resistant to chemotherapy. Other references illustrating the use of

retroviral vectors in gene therapy are: Clowes et al., J. Clin. Invest. 93:644-
651
(1994); Kiem et al., Blood 83:1467-1473 (1994); Salmons and Gunzberg,
Human Gene Therapy 4:129-141 (1993); and Grossman and Wilson, Curr.
Opin. in Genetics and Devel. 3:110-114 (1993).
[0386] Adenoviruses are other viral vectors that can be used in gene
therapy.
Adenoviruses are especially attractive vehicles for delivering genes to
respiratory epithelia. Adenoviruses naturally infect respiratory epithelia
where
they cause a mild disease. Other targets for adenovirus-based delivery systems

are liver, the central nervous system, endothelial cells, and muscle.
Adenoviruses have the advantage of being capable of infecting non-dividing
cells. Kozarsky and Wilson, Current Opinion in Genetics and Development
3:499-503 (1993) present a review of adenovirus-based gene therapy. Bout et
al., Human Gene Therapy 5:3-10 (1994) demonstrated the use of adenovirus
vectors to transfer genes to the respiratory epithelia of rhesus monkeys.
Other
instances of the use of adenoviruses in gene therapy can be found in Rosenfeld

et al., Science 252:431434 (1991); Rosenfeld et al., Cell 68:143-155 (1992);
Mastrangeli et al., J. Clin. Invest. 91:225-234 (1993); PCT Publication
W094/12649; and Wang, et al., Gene Therapy 2:775-783 (1995). In a
preferred embodiment, adenovirus vectors are used.
[0387] Adeno-associated virus (AAV) has also been proposed for use in gene
therapy (Walsh et al., Proc. Soc. Exp. Biol. Med. 204:289-300 (1993); U.S.
Pat. No. 5,436,146).
[0388] Another approach to gene therapy involves transferring a gene to
cells
in tissue culture by such methods as electroporation, lipofection, calcium
phosphate mediated transfection, or viral infection. Usually, the method of
transfer includes the transfer of a selectable marker to the cells. The cells
are

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
141
then placed under selection to isolate those cells that have taken up and are
expressing the transferred gene. Those cells are then delivered to a patient.
[0389] In this embodiment, the nucleic acid is introduced into a cell
prior to
administration in vivo of the resulting recombinant cell. Such introduction
can
be carried out by any method known in the art, including but not limited to
transfection, electroporation, microinjection, infection with a viral or
bacteriophage vector containing the nucleic acid sequences, cell fusion,
chromosome-mediated gene transfer, microcell-mediated gene transfer,
spheroplast fusion, etc. Numerous techniques are known in the art for the
introduction of foreign genes into cells (see, e.g., Loeffler and Behr, Meth.
Enzymol. 217:599-618 (1993); Cohen et al., Meth. Enzymol. 217:618-644
(1993); Cline, Pharmac. Ther. 29:69-92m (1985) and may be used in
accordance with the present invention, provided that the necessary
developmental and physiological functions of the recipient cells are not
disrupted. The technique should provide for the stable transfer of the nucleic

acid to the cell, so that the nucleic acid is expressible by the cell and
preferably
heritable and expressible by its cell progeny.
[0390] The resulting recombinant cells can be delivered to a patient by
various
methods known in the art. Recombinant blood cells (e.g., hematopoietic stem
or progenitor cells) are preferably administered intravenously. The amount of
cells envisioned for use depends on the desired effect, patient state, etc.,
and
can be determined by one skilled in the art.
[0391] Cells into which a nucleic acid can be introduced for purposes of
gene
therapy encompass any desired, available cell type, and include but are not
limited to epithelial cells, endothelial cells, keratinocytes, fibroblasts,
muscle
cells, hepatocytes; blood cells such as Tlymphocytes, Blymphocytes,
monocytes, macrophages, neutrophils, eosinophils, megakaryocytes,
granulocytes; various stern or progenitor cells, in particular hematopoietic
stem or progenitor cells, e.g., as obtained from bone marrow, umbilical cord
blood, peripheral blood, fetal liver, etc.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
142
[0392] In a preferred embodiment, the cell used for gene therapy is
autologous
to the patient.
[0393] In an embodiment in which recombinant cells are used in gene
therapy,
nucleic acid sequences encoding an antibody are introduced into the cells such

that they are expressible by the cells or their progeny, and the recombinant
cells are then administered in vivo for therapeutic effect. In a specific
embodiment, stem or progenitor cells are used. Any stem and/or progenitor
cells which can be isolated and maintained in vitro can potentially be used in

accordance with this embodiment of the present invention (see e.g. PCT
Publication WO 94/08598; Stemple and Anderson, Cell 71:973-985 (1992);
Rheinwald, Meth. Cell Bio. 21A:229 (1980); and Pittelkow and Scott, Mayo
Clinic Proc. 61:771 (1986)).
[0394] In a specific embodiment, the nucleic acid to be introduced for
purposes of gene therapy comprises an inducible promoter operably linked to
the coding region, such that expression of the nucleic acid is controllable by

controlling the presence or absence of the appropriate inducer of
transcription.
Hyperproliferative Diseases
[0395] The method of the invention, can be used to treat
hyperproliferative
diseases, disorders, and/or conditions, including neoplasms.
[0396] Examples of hyperproliferative diseases, disorders, and/or
conditions
that can be treated by the method of the invention include, but are not
limited
to neoplasms located in the: prostate, colon, abdomen, bone, breast, digestive

system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid,
pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous
(central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen,

thoracic, and urogenital.
[0397] Other examples of such hyperproliferative disorders include, but
are
not limited to: Acute Childhood Lymphoblastic Leukemia, Acute
Lymphoblastic Leukemia, Acute Lymphocytic Leukemia, Acute Myeloid

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
143
Leukemia, Adreno cortical Carcinoma, Adult (Primary) Hepatocellular Cancer,
Adult (Primary) Liver Cancer, Adult Acute Lymphocytic Leukemia, Adult
Acute Myeloid Leukemia, Adult Hodgkin's Disease, Adult Hodgkin's
Lymphoma, Adult Lymphocytic Leukemia, Adult Non-Hodgkin's Lymphoma,
Adult Primary Liver Cancer, Adult Soft Tissue Sarcoma, AIDS-Related
Lymphoma, AIDS-Related Malignancies, Anal Cancer, Astrocytoma, Bile
Duct Cancer, Bladder Cancer, Bone Cancer, Brain Stem Glioma, Brain
Tumors, Breast Cancer, Cancer of the Renal Pelvis and Ureter, Central
Nervous System (Primary) Lymphoma, Central Nervous System Lymphoma,
Cerebellar Astrocytoma, Cerebral Astrocytoma, Cervical Cancer, Childhood
(Primary) Hepatocellular Cancer, Childhood (Primary) Liver Cancer,
Childhood Acute Lymphoblastic Leukemia, Childhood Acute Myeloid
Leukemia, Childhood Brain Stem Glioma, Childhood Cerebellar Astrocytoma,
Childhood Cerebral Astrocytoma, Childhood Extracranial Germ Cell Tumors,
Childhood Hodgkin's Disease, Childhood Hodgkin's Lymphoma, Childhood
Hypothalamic and Visual Pathway Glioma, Childhood Lymphoblastic
Leukemia, Childhood Medulloblastoma, Childhood Non-Hodgkin's
Lymphoma, Childhood Pineal and Supratentorial Primitive Neuroectodermal
Tumors, Childhood Primary Liver Cancer, Childhood Rhabdomyo sarcoma,
Childhood Soft Tissue Sarcoma, Childhood Visual Pathway and Hypothalamic
Glioma, Chronic Lymphocytic Leukemia, Chronic Myelogenous Leukemia,
Colon Cancer, Cutaneous T-Cell Lymphoma, Endocrine Pancreas Islet Cell
Carcinoma, Endometrial Cancer, Ependymoma, Epithelial Cancer, Esophageal
Cancer, Ewing's Sarcoma and Related Tumors, Exocrine Pancreatic Cancer,
Extracranial Germ Cell Tumor, Extragonadal Germ Cell Tumor, Extrahepatic
Bile Duct Cancer, Eye Cancer, Female Breast Cancer, Gaucher's Disease,
Gallbladder Cancer, Gastric Cancer, Gastrointestinal Carcinoid Tumor,
Gastrointestinal Tumors, Germ Cell Tumors, Gestational Trophoblastic
Tumor, Hairy Cell Leukemia, Head and Neck Cancer, Hepatocellular Cancer,
Hodgkin's Disease, Hodgkin's Lymphoma, Hypergammaglobulinemia,
Hypopharyngeal Cancer, Intestinal Cancers, Intraocular Melanoma, Islet Cell

CA 02548180 2006-06-01
WO 2005/055936 PCT/US2004/040573
144
Carcinoma, Islet Cell Pancreatic Cancer, Kaposi's Sarcoma, Kidney Cancer,
Laryngeal Cancer, Lip and Oral Cavity Cancer, Liver Cancer, Lung Cancer,
Lymphoproliferative Disorders, Macroglobulinemia, Male Breast Cancer,
Malignant Mesothelioma, Malignant Thymoma, Medulloblastoma, Melanoma,
Mesothelioma, Metastatic Occult Primary Squamous Neck Cancer, Metastatic
Primary Squamous Neck Cancer, Metastatic Squamous Neck Cancer, Multiple
Myeloma, Multiple Myeloma/Plasma Cell Neoplasm, Myelodysplastic
Syndrome, Myelogenous Leukemia, Myeloid Leukemia, Myeloproliferative
Disorders, Nasal Cavity and Paranasal Sinus Cancer, Nasopharyngeal Cancer,
Neuroblastoma, Non-Hodgkin's Lymphoma During Pregnancy, Nonmelanoma
Skin Cancer, Non-Small Cell Lung Cancer, Occult Primary Metastatic
Squamous Neck Cancer, Oropharyngeal Cancer, Osteo-/Malignant Fibrous
Sarcoma, Osteosarcoma/Malignant Fibrous Histiocytoma,
Osteosarcoma/Malignant Fibrous Histiocytoma of Bone, Ovarian Epithelial
Cancer, Ovarian Germ Cell Tumor, Ovarian Low Malignant Potential Tumor,
Pancreatic Cancer, Paraproteinemias, Purpura, Parathyroid Cancer, Penile
Cancer, Pheochromocytoma, Pituitary Tumor, Plasma Cell Neoplasm/Multiple
Myeloma, Primary Central Nervous System Lymphoma, Primary Liver
Cancer, Prostate Cancer, Rectal Cancer, Renal Cell Cancer, Renal Pelvis and
Ureter Cancer, Retinoblastoma, Rhabdomyosarcoma, Salivary Gland Cancer,
Sarcoidosis Sarcomas, Sezary Syndrome, Skin Cancer, Small Cell Lung
Cancer, Small Intestine Cancer, Soft Tissue Sarcoma, Squamous Neck Cancer,
Stomach Cancer, Supratentorial Primitive Neuroectodermal and Pineal
Tumors, T-Cell Lymphoma, Testicular Cancer, Thymoma, Thyroid Cancer,
Transitional Cell Cancer of the Renal Pelvis and Ureter, Transitional Renal
Pelvis and Ureter Cancer, Trophoblastic Tumors, Ureter and Renal Pelvis Cell
Cancer, Urethral Cancer, Uterine Cancer, Uterine Sarcoma, Vaginal Cancer,
Visual Pathway and Hypothalamic Glioma, Vulvar Cancer, Waldenstrom's
Macroglobulinemia, Wilms' Tumor, and any other hyperproliferative disease,
besides neoplasia, located in an organ system listed above.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
145
[0398] The method of the present invention may be used to treat
premalignant
conditions and to prevent progression to a neoplastic or malignant state,
including but not limited to those disorders described above. Such uses are
indicated in conditions known or suspected of preceding progression to
neoplasia or cancer, in particular, where non-neoplastic cell growth
consisting
of hyperplasia, metaplasia, or most particularly, dysplasia has occurred (for
review of such abnormal growth conditions, see Robbins and Angell, 1976,
Basic Pathology, 2d Ed., W. B. Saunders Co., Philadelphia, pp. 68-79.)
[0399] Hyperplasia is a form of controlled cell proliferation, involving
an
increase in cell number in a tissue or organ, without significant alteration
in
structure or function. Hyperplastic disorders which can be treated by the
method of the invention include, but are not limited to, angiofollicular
mediastinal lymph node hyperplasia, angiolymphoid hyperplasia with
eosinophilia, atypical melanocytic hyperplasia, basal cell hyperplasia, benign

giant lymph node hyperplasia, cementum hyperplasia, congenital adrenal
hyperplasia, congenital sebaceous hyperplasia, cystic hyperplasia, cystic
hyperplasia of the breast, denture hyperplasia, ductal hyperplasia,
endometrial
hyperplasia, fibromuscular hyperplasia, focal epithelial hyperplasia, gingival

hyperplasia, inflammatory fibrous hyperplasia, inflammatory papillary
hyperplasia, intravascular papillary endothelial hyperplasia, nodular
hyperplasia of prostate, nodular regenerative hyperplasia,
pseudoepitheliomatous hyperplasia, senile sebaceous hyperplasia, and
verrucous hyperplasia.
[0400] Metaplasia is a form of controlled cell growth in which one type of
adult or fully differentiated cell substitutes for another type of adult cell.

Metaplastic disorders which can be treated by the method of the invention
include, but are not limited to, agnogenic myeloid metaplasia, apocrine
metaplasia, atypical metaplasia, autoparenchymatous metaplasia, connective
tissue metaplasia, epithelial metaplasia, intestinal metaplasia, metaplastic
anemia, metaplastic ossification, metaplastic polyps, myeloid metaplasia,
primary myeloid metaplasia, secondary myeloid metaplasia, squamous

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
146
metaplasia, squamous metaplasia of amnion, and symptomatic myeloid
metaplasia.
[04011 Dysplasia is frequently a forerunner of cancer, and is found mainly
in
the epithelia; it is the most disorderly form of non-neoplastic cell growth,
involving a loss in individual cell uniformity and in the architectural
orientation of cells. Dysplastic cells often have abnormally large, deeply
stained nuclei, and exhibit pleomorphism. Dysplasia characteristically occurs
where there exists chronic irritation or inflammation. Dysplastic disorders
which can be treated by the method of the invention include, but are not
limited to, anhidrotic ectodermal dysplasia, anterofacial dysplasia,
asphyxiating thoracic dysplasia, atriodigital dysplasia, bronchopulmonary
dysplasia, cerebral dysplasia, cervical dysplasia, chondroectodermal
dysplasia,
cleidocranial dysplasia, congenital ectodermal dysplasia, craniodiaphysial
dysplasia, craniocarpotarsal dysplasia, craniometaphysial dysplasia, dentin
dysplasia, diaphysial dysplasia, ectodermal dysplasia, enamel dysplasia,
encephalo-ophthalmic dysplasia, dysplasia epiphysialis hemimelia, dysplasia
epiphysialis multiplex, dysplasia epiphysialis punctata, epithelial dysplasia,

faciodigitogenital dysplasia, familial fibrous dysplasia of jaws, familial
white
folded dysplasia, fibromuscular dysplasia, fibrous dysplasia of bone, florid
osseous dysplasia, hereditary renal-retinal dysplasia, hidrotic ectodermal
dysplasia, hypohidrotic ectodermal dysplasia, lymphopenic thymic dysplasia,
mammary dysplasia, mandibulofacial dysplasia, metaphysial dysplasia,
Mondini dysplasia, mono stotic fibrous dysplasia, mucoepithelial dysplasia,
multiple epiphysial dysplasia, oculoauriculovertebral dysplasia,
oculodentodigital dysplasia, oculovertebral dysplasia, odontogenic dysplasia,
ophthalmomandibulomelic dysplasia, periapical cemental dysplasia,
polyostotic fibrous dysplasia, pseudoachondroplastic spondyloepiphysial
dysplasia, retinal dysplasia, septo-optic dysplasia, spondyloepiphysial
dysplasia, and ventriculoradial dysplasia.
[04021 Additional pre-neoplastic disorders which can be treated by the
method
of the invention include, but are not limited to, benign dysproliferative

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
147
disorders (e.g., benign tumors, fibrocystic conditions, tissue hypertrophy,
intestinal polyps, colon polyps, and esophageal dysplasia), leukoplakia,
keratoses, Bowen's disease, Farmer's Skin, solar cheilitis, and solar
keratosis.
[0403] In preferred embodiments, the method of the invention is used to
inhibit growth, progression, and/or metastasis of cancers, in particular those

listed above.
[0404] Additional diseases or conditions associated with increased cell
survival that could be treated by the method of the invention, include, but
are
not limited to, progression, and/or metastases of malignancies and related
disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic

leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic,
myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias
(e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic
leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-
Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia,
heavy chain disease, and solid tumors including, but not limited to, sarcomas
and
carcinomas such as fibrosarcoma, myxo sarcoma, lip osarcoma,
chondrosarcoma, osteogenic sarcoma, chordoma,
angio sarcoma,
endotheliosarcoma, lymphangio sarcoma, lymphangioendothelio sarcoma,
synovioma, mesothelioma, Ewing's tumor,
leiomyosarcoma,
rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian
cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma,
adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary
carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary
carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct
carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor,
cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma,
bladder carcinoma, epithelial carcinoma, glioma, astrocytoma,
medulloblastoma, craniopharyngioma, ependymoma,
pinealoma,
emangioblastoma, acoustic neuroma, oligodendroglioma, menangioma,
melanoma, neuroblastoma, and retinoblastoma.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
148
[0405] Hyperproliferative diseases and/or disorders that could be treated
by
the method of the invention, include, but are not limited to, neoplasms
located
in the liver, abdomen, bone, breast, digestive system, pancreas, peritoneum,
endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus,
thyroid), eye, head and neck, nervous system (central and peripheral),
lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and urogenital
tract.
[0406] Similarly, other hyperproliferative disorders can also be treated
by the
method of the invention. Examples of such hyperproliferative disorders
include, but are not limited to: hypergammaglobulinemia, lymphoproliferative
disorders, paraproteinemias, purpura, sarcoidosis, Sezary Syndrome,
Waldenstron's macroglobulinemia, Gaucher's Disease, histiocytosis, and any
other hyperproliferative disease, besides neoplasia, located in an organ
system
listed above.
Demonstration Of Therapeutic Activity
[0407] The methods and antibodies of the invention are preferably tested
in
vitro, and then in vivo for the desired therapeutic or prophylactic activity,
prior
to use in humans. For example, in vitro assays to demonstrate the therapeutic
or prophylactic utility of a compound or pharmaceutical composition include
the effect of a compound on a cell line or a patient tissue sample. The effect
of
the compound or composition on the cell line and/or tissue sample can be
determined utilizing techniques known to those of skill in the art including,
but
not limited to, rosette formation assays and cell lysis assays. In accordance
with the invention, in vitro assays which can be used to determine whether
administration of a specific compound is indicated, include in vitro cell
culture
assays in which a patient tissue sample is grown in culture, and exposed to or

otherwise administered a compound, and the effect of such compound upon
the tissue sample is observed.
Kits

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
149
[0408] The present invention provides kits that can be used in the above
methods. In one embodiment, a kit comprises an antibody of the invention,
preferably a purified antibody, in one or more containers. In a specific
embodiment, the kits of the present invention contain a substantially isolated

polypeptide comprising an epitope which is specifically immunoreactive with
an antibody included in the kit. Preferably, the kits of the present invention

further comprise a control antibody which does not react with the polypeptide
of interest. In another specific embodiment, the kits of the present invention

contain a means for detecting the binding of an antibody to a polypeptide of
interest (e.g., the antibody may be conjugated to a detectable substrate such
as
a fluorescent compound, an enzymatic substrate, a radioactive compound or a
luminescent compound, or a second antibody which recognizes the first
antibody may be conjugated to a detectable substrate).
[0409] In another specific embodiment of the present invention, the kit is
a
diagnostic kit for use in screening serum containing antibodies specific
against
proliferative and/or cancerous polynucleotides and polypeptides. Such a kit
may include a control antibody that does not react with the polypeptide of
interest. Such a kit may include a substantially isolated polypeptide antigen
comprising an epitope which is specifically immunoreactive with at least one
anti-polypeptide antigen antibody. Further, such a kit includes means for
detecting the binding of said antibody to the antigen (e.g., the antibody may
be
conjugated to a fluorescent compound such as fluorescein or rhodamine which
can be detected by flow cytometry). In specific embodiments, the kit may
include a recombinantly produced or chemically synthesized polypeptide
antigen. The polypeptide antigen of the kit may also be attached to a solid
support.
[0410] In a more specific embodiment the detecting means of the above-
described kit includes a solid support to which said polypeptide antigen is
attached. Such a kit may also include a non-attached reporter-labeled anti-
human antibody. In this embodiment, binding of the antibody to the

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
150
polypeptide antigen can be detected by binding of the said reporter-labeled
antibody.
[0411] In an additional embodiment, the invention includes a diagnostic
kit for
use in screening samples containing antigens of the polypeptide of the
invention. The diagnostic kit includes a substantially isolated antibody
specifically immunoreactive with polypeptide or polynucleotide antigens, and
means for detecting the binding of the polynucleotide or polypeptide antigen
to
the antibody. In one embodiment, the antibody is attached to a solid support.
In
a specific embodiment, the antibody may be a monoclonal antibody. The
detecting means of the kit may include a second, labeled monoclonal antibody.
Alternatively, or in addition, the detecting means may include a labeled,
competing antigen.
[0412] In one diagnostic configuration, test sample is reacted with a
solid
phase reagent having a surface-bound antigen obtained by the methods of the
present invention. After binding with specific antigen antibody to the reagent

and removing unbound sample components by washing, the reagent is reacted
with reporter-labeled anti-human antibody to bind reporter to the reagent in
proportion to the amount of bound anti-antigen antibody on the solid support.
The reagent is again washed to remove unbound labeled antibody, and the
amount of reporter associated with the reagent is determined. Typically, the
reporter is an enzyme which is detected by incubating the solid phase in the
presence of a suitable fiuorometric, luminescent or calorimetric substrate
(Sigma, St. Louis, Mo.).
[0413] The solid surface reagent in the above assay is prepared by known
techniques for attaching protein material to solid support material, such as
polymeric beads, dip sticks, 96-well plate or filter material. These
attachment
methods generally include non-specific adsorption of the protein to the
support
or covalent attachment of the protein, typically through a free amine group,
to
a chemically reactive group on the solid support, such as an activated
carboxyl,
hydroxyl, or aldehyde group. Alternatively, streptavidin coated plates can be
used in conjunction with biotinylated antigen(s).

CA 02548180 2011-12-28
/151
[0414] Thus, the
invention provides an assay system or kit for carrying out this
diagnostic method. The kit generally includes a support with surface- bound
recombinant antigens, and a reporter-labeled anti-human antibody for
detecting surface-bound anti-antigen antibody.
Apoptosis-Inducing Therapies
[0415] Apoptosis-
inducing therepies include chemotherapeutic agents (also
known as antineoplastic agents), radiation therapy, and combination
radiotherapy and chemotherapy.
[0416] Exemplary chemotherapeutic agents are vinca alkaloids,
epipodophyllotoxins, anthracycline antibiotics, actinomycin D, plicamycin,
TM
puromycin, gramicidin D, paclitaxel (Taxo1 t Bristol Myers
Squibb),
colchicine, cytochalasin B, emetine, maytansine, and amsacrine (or
"m_AMSA"). The vinca alkaloid class is described in Goodman and Gilman's
The Pharmacological Basis of Therapeutics (7th ed.), (1985), pp. 1277-1280.
Exemplary of vinca alkaloids are vincristine, vinblastine, and vindesine. The
epipodophyllotoxin class is described in Goodman and Gilman's The
Pharmacological Basis of Therapeutics (7th ed.), (1985), pp. 1280-1281.
Exemplary of epipodophyllotoxins are etoposide, etoposide orthoquinone, and
teniposide. The anthracycline antibiotic class is described in Goodman and
Gilman's The Pharmacological Basis of Therapeutics (7th ed.), (1985), pp.
1283-1285. Exemplary of anthracycline antibiotics are daunorubicin,
doxorubicin, mitoxantraone, and bisanthrene. Actinomycin D, also called
Dactinomycin, is described in Goodmand and Gilman's The Pharmacological
Basis of Therapeutics (7th ed.), (1985), pp. 1281-1283. Plicamycin, also
called
mithramycin, is described in Goodmand and Gilman's The Pharmacological
Basis of Therapeutics (7th ed), (1985), pp .1287-1288. Additional
chemotherapeutic agents include cisplatin (Platino
= im Bristol
Myers
TM
Squibb), carboplatin (Paraplatin, Bristol Myers
Squibb), mitomycin
TM TM
(Mutamycin, Bristol Myers Squibb),
altretamine (Hexalen, U.S.

CA 02548180 2011-12-28
152
TM
Bioscience, Inc.), cyclophospharnide (Cytoxan, Bristol Myers
Squibb),
TM
lomustine (CCNU) (CeeNU, Bristol Myers
Squibb), carmustine
TM
(BCNU) (BiCNU, Bristol Myers Squibb).
[0417] Exemplary chemotherapeutic agents also include aclacinomycin A,
aclambicin, acronine, acronycine, adriamycin, aldesleukin (interleukin-2),
altretamine (hexamiethylmelamine), aminoglutethimide, aminoglutethimide
(cytada), aminoimidazole carboxamide, amsacrine (m-A_MSA; amsidine),
anastrazole (arimida), ancitabine, anthracyline, anthrarnycin, asparaginase
(elspar), azacitdine, azacitidine (ladakamycin)T,m azaguanine, azaserine,
azauridine, 1,11,1"-phosphinothioylidynetris aziridine,
azirino(2',
3':3,4)pyrrolo(1,2-a)indole-4,7-dione, BCG (theraclY1), BCNU, BCNU
chloroethyl nitrosoureas, benzamide, 4-(bis(2-
chloro ethyl)amino)benzenebutanoic acid, bicalutamide, bischloro ethyl
TM
nitrosourea, bleomycin, bleomycin (blenozane), bleomycins,
bromodeoxyuridine, broxuridine, busulfan (mylea), carbamic acid ethyl
TM
ester, carboplatin, carboplatin (paraplatin), carmustine, carmustine (BCNU;
TM
BiCNU), chlorambucil (leukeran), chloroethyl nitrosoureas, chorozotocin
TM
(DCNU), chromomycin A3, cis-retinoic acid, cisplatin (cis-ddpl; platinol),
TM
c lacirib ine (2-chlorodeoxyadenosine; 2cda; leustatin),
cofonnycin,
cycloleucine, cyclophosphamide, cyclophosphamide anhydrous, chlorambucil,
TM
cytarabine, cytarabine, cytarabine HC1 (cytosar-u), 2-deoxy-2-
(((methyhiitrosoamino)carbonyl)amino)-D-glucose, dacarbazine, dactinomycin
(cosmegeri), daunorubicin, Daunorubincin HC1 (cerubidia decarbazine,
decarbazine (DTIC-do), demecolcine, dexamethasone, dianhycirogalactitol,
Tm
diazooxonorleucine, diethylstilbestrol, docetaxel (taxotere), doxorubicin HC1
(adriamycizij, doxorubicin hydrochloride, eflomithine, estramustine,
estramustine phosphate sodium (emcyt), ethiodized oil, ethoglucid, ethyl
carbarnate, ethyl methanesulfonate, etoposide (VP16-213), fenretinide,
TM TM TM
floxtuidine, floxuridine (fudr), fludarabine (fludara), fluorouracil (5-FU),
TM TM
fluoxymesterone (halotestm), flutamide, flutamide (eulexin), fluxuridine,
TM
gallium nitrate (granite), gemcitabine (gemzg), genistein, 2-deoxy-2-(3-
_

CA 02548180 2011-12-28
153
methyl-3-nitrosoureido)-D-glucopyranose, goserelin (zoladj)), hexestrol,
hydroxyurea (hydr71), idarubicin (idamycifil ifosfagemcitabine, ifosfamide
(ifiex), ifosfamide with mesna (MAID), interferon, interferon alfa, interferon

alfa-2a, alfa-2b, alfa-n3, interleukin-2, iobenguane, iobenguane iobenguane,
irinotecan (camptosTr), isotretinoin (accutahme), ketoconazole, 4-(bis(2-
chloroethyl)amino)-L-phenylalanine, L-serine diazoacetate, lentinan,
leucovorin, leuprolide acetate (LH:RH-analog), levamisole (ergamisTol),
TM
lomustine (CCNU; cee-NU), marmomustine, maytansine, mechlorethamine,
mechlorethamine HC1 (nitrogen mustarn4d), medroxyprogesterone acetate
TM TM
(proverTMa, dcipo prove), megestrol acetate (menace), melengestrol acetate,
TM TM
melphalan (alkeran), menogaril, mercaptopmin, mercaptopurine (purinethol),
mercaptopurine anhydrous, MESNA, mesna (mesa), methanesulfonic acid,
=
ethyl ester, methotrexate methotrexate), methyl-ccnu, mimo sine,
misonidazole, mithramycin, mitoantrone, mitobronitol, mitoguazone,
TM =
mitolactol, mitomycin (mutamycin), mitomycin C, mitotane (o,p'-DDD;
lysodren), mitoxantrone, mitoxantrone HC1 (novantrolie), mopidamol, N,N-
bis(2-chloroethyl)tetrahydro-2H-1,3,2-oxazaphosphorin-2-amine-2-oxide, N-
(1-methylethyl)-4-((2-methylhydrazin.o)methyl)benzamide, N-methyl-bis(2-
chIoroethypamine, nicardipine, nilutamide (nilandroTrim), nimustine,
nitracrine,
TM
nitrogen mustard, nocodazole, nogalarnycin, octreotide (sandostatin),
TM TM
pacilataxel (taxon), paclitaxel, pactamycin, pegaspargase (PEGx-1),
pentostatin (2'-deoxycoformycin), peplomycin, peptichemio, photophoresis,
picamycin (mithracijr, picibanil, pipobroman, plicamycin, podofilox,
podophyllotoxin, porfiromycin, prednisone, procarbazine, procarbazine HC1
(matulake), prospidium, puromycin, puromycin aminonucleoside, PUVA
(psoralen+ultraviolet a), pyran copolymer, rapamycin, s-azacytidine, 2,4,6-
tris(1-aziridiny1)-s-triazine, semustine, showdomycin, sirolimus, streptozocin
TM fl4 TM TMTM
(zanosar), surarnin, tamoxifen citrate (nolvadex), taxon, tegafur, teniposide
(VM-26; wunTomn), tenuazonic acid, TEPA, testolactone, thio-tepa, thioguanine,
TM Tm
thiotepa (thioplex), tilorone, topotecan, tretinoin (vesanoid), triaziquone,
trichodermin, triethylene glycol diglycidyl ether, triethylenemelamine,

CA 02548180 2011-12-28
154
triethylenephosphoramide, triethylenethiophosphoramide,
trimetrexate
TM
(neutrexin), tris(1-aziridinyl)phosphine oxide, tris(1-aziridinyl)phosphine
sulfide, tris(aziridinyI)-p-benzoquinone, tris(aziridinyl)phosphine sulfide,
uracil mustard, vidarabine, vidarabine phosphate, vinblastine, vinblastine
TM TM
sulfate (velban), vincristine sulfate (oncovin), vindesine, vinorelbine,
TM
vinorelbine tartrate (navelbine), (1)-mimosine, 1-(2-chloroethyl)-3-(4-
methylcyclohexyl)-1 -nitro sourea, (8S-cis)-10-((3-amino-2,3,6-trideoxy-alpha-
L-lyxo-hexopyranosyl)oxy)-7,8,9 ,10-tetrahydro-6,8, 11-trihydroxy-8-
(hydroxyacety1)-1-methoxy-5,12-naphthacenedione, 131 -meta-iodob
enzyl
guanidine (1-131 MIBG), 5-(3,3-dimethy1-1-triazeny1)-1H-imidazole-4-
carboxamide, 5-(bis(2-chloroethyl)amino)-2,4(1H,3H)-pyrimidinedione, 2,4,6-
tris(1-aziri diny1)-s-thia.zine, 2,3,5-tris(1-
aziridiny1)-2,5-cyclohexadiene-1,4-
dione, 2-chloro-N-(2-chloroethyl)-N-
methylethanamine, N,N-bis(2-
chloroethyl)tetrahydro-2H-1,3 ,2-ox azapho sphorin-2-amine-2-oxi de, 3-
deazauridine, 3-iodobenzylguanidine, 5,12-naphthacenedione, 5-azacytidine,
5-fluorouracil, (1 aS,8 S ,8 aR,8b S)-6-amino-8-(((aminocarbonypoxy)methyl)-
1,1 a, 2,8,8 a,8b-hexahydro-8 a-methoxy-5-methylazirino(2',3% 3,4)pyrrolo (1,2-

a)indole-4,7-dione, 6-azauridine, 6-mercaptopurine, 8-azaguanine, and
combinations thereof.
[0418] Preferred therapeutic agents and combinations that may be
administered as an apoptosis-inducing therapy include Doxorubicin and
Doxetaxel, Topotecan, Paclitaxel, Carboplatin and Taxol, Taxol, Cisplatin and
Radiation, 5-fluorouracil (5-FU), 5-FU and Radiation, Toxotere, Fludarabine,
Ara C, Etoposide, Vincristine, and Vinblastin.
[0419] Exemplary
chemotherapeutic agents also include doxetaxel
TM TM
(TOXOTERE) and to potecan (HYCAMTIN).
[0420] Chemotherapeutic agents that may be administered in the method of
the invention include, but are not limited to, antibiotic derivatives (e.g.,
doxorubicin, bleomycin, daunorubicin, and dactinomycin); antiestrogens (e.g.,
tamoxifen); antimetabolites (e.g., fluorouracil, 5-FU, methotrexate,
floxuridine, interferon alpha-2b, glutamic acid, plicamycin, mercaptopurine,

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
155
and 6-thioguanine); cytotoxic agents (e.g., carmustine, BCNU, lomustine,
CCNU, cytosine arabinoside, cyclophosphamide, estramustine, hydroxyurea,
procarbazine, mitomycin, busulfan, cis-platin, and vincristine sulfate);
hormones (e.g., medroxyprogesterone, estramustine phosphate sodium, ethinyl
estradiol, estradiol, megestrol acetate, methyltestosterone,
diethylstilbestrol
diphosphate, chlorotrianisene, and testolactone); nitrogen mustard derivatives

(e.g., mephalen, chorambucil, mechlorethamine (nitrogen mustard) and
thiotepa); steroids and combinations (e.g., bethamethasone sodium phosphate);
and others (e.g., dicarbazine, asparaginase, mitotane, vincristine sulfate,
vinblastine sulfate, and etoposide).
[0421] In a specific embodiment, antibodies of the invention are
administered
in combination with CHOP (cyclophosphamide, doxorubicin, vincristine, and
prednisone) or any combination of the components of CHOP. In another
embodiment, antibodies of the invention are administered in combination with
Rituximab. In a further embodiment, antibodies of the invention are
administered with Rituxmab arid CHOP, or Rituxmab and any combination of
the components of CHOP.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
156
Table 4: Commonly Used Chemotherapy Drugs for Major Cancer Indications
1. Breast cancer: Adjuvant therapy (systemic therapy as an adjunct to or in
addition to surgery). Doxorubicin (Adriamycin), cyclophosphamide, and
taxanes [paclitaxel (Taxol) and docetaxel (Taxotere)]. These three drugs
are also active in metastatic breast cancer but if the patient has already
received them as adjuvant therapy the commonly used drugs are
capecitabine (Xeloda), gemcitabine (Gemzar), vinorelbine (Navelbine).
Commonly prescribed hormonal agents for bone metastases of hormone
receptor positive tumors are: tamoxifen and aromatase inhibitors
(Arimidex, Femara, Aromasin).
2. Colon cancer: 5-FU plus leucovorin, irinotecan (camptosar), oxaliplatin,
and capecitabine.
3. Lung cancer: Cisplatin, carboplatin, paclitaxel, docetaxel, gemcitabine,
vinorelbine.
4. Prostate cancer: Docetaxel, estramustine, mitoxantrone (Novantrone),
and prednisone.
5. Non-Hodgkin's Lymphoma: Cyclophosphamide, doxorubicin,
vincristine (Oncovin), and prednisone.
[0422] Therapeutic radiation includes, for example, fractionated
radiotherapy,
nonfractionated radiotherapy and hyperfractionated radiotherapy, and
combination radiation and chemotherapy. Types of radiation also include
ionizing (gamma) radiation, particle radiation, low energy transmission (LET),

high energy transmission (HET), ultraviolet radiation, infrared radiation,
visible light, and photosensitizing radiation. As used herein, chemotherapy
includes treatment with a single chemotherapeutic agent or with a combination
of agents. In a subject in need of treatment, chemotherapy may be combined
with surgical treatment or radiation therapy, or with other antineoplastic
treatment modalities.

CA 02548180 2011-12-28
157
Other Therapeutics
104231 In a further
embodiment, the antibodies of the invention are
administered in combination with an antiviral agent. Antiviral agents that may

be administered with the antibodies of the invention include, but are not
limited to, acyclovir, ribavirin, amantadine, and remantidine.
[04241 Antibodies of the
invention may also be administered with antiemetics
such as 2-(ethylthio)-1 0-(3 -(4-methyl- 1 -pip era.zinyl)
propy1)- 1 OH-
phenothiazine (ethylthioperazine), 1-(p-chloro-alpha-phenylbenzy1)-4-(m-
TM TM
methylbenzyp-piperazine (meclozine, meclizine), etc., and combinations
thereof. Polynucleotides and polypeptides of the invention may also be
administered with other therapeutic agents, and combinations thereof,
disclosed herein or known in the art.
[04251 In a further embodiment, the antibodies of the invention are
administered in combination with an antibiotic agent. Antibiotic agents that
may be administered with the antibodies of the invention include, but are not
limited to, amoxicillin, beta-lactamases, aminoglycosides, beta-lactam
(glycopeptide), beta-lactamase,s, Clindamycin, chloramphenicol,
cephalosporins, ciprofloxacin, ciprofloxacin, erythromycin, fluoroquinolones,
macrolides, metronidazole, penicillins, quinolones, rifampin, streptomycin,
sulfonamide, tetracyclines, trimethoprim, trimethoprim-sulfamthoxazole, and
vancomycin.
[0426] Conventional nonspecific inununosuppressive agents, that may be
administered in combination with the antibodies of the invention include, but
are not limited to, steroids, cyclosporine, cyclosporine analogs,
cyclophosphamide methylprednisone, prednisone, azathioprine, FK-506, 15-
deoxyspergualin, and other immunosuppressive agents that act by suppressing
the function of responding T cells.
[04271 In specific embodiments, antibodies of the invention are
administered
in combination with immunosuppressants. Immunosuppressants preparations
that may be administered with the antibodies of the invention include, but are

CA 02548180 2011-12-28
158
TM
not limited to, ORTHOCLONE (OKT3),
TM TM TM
SANDIMMUNE /NEORA1 =/SANGDYA (cyclosporin),
TM TM
PROGRAF (tacrolimus), CELLCEPT (mycophenolate),
TM
Azathioprine, glucorticosteroids, and RAPAMUNE (sirolimus). In
a
specific embodiment, immunosuppressants may be used to prevent rejection of
organ or bone marrow transplantation.
[0428] In an additional embodiment, antibodies of the invention are
administered alone or in combination with one or more intravenous immune
globulin preparations. Intravenous immune globulin preparations that may be
administered with the antibodies of the invention include, but not limited to,
TM
TM
TM
GAMMAR) IVEEGAM SANDOGLOBULIN;
TM TM
GAMMAGARD S/D and GAMIMUNE.' In a specific
embodiment,
antibodies of the invention are administered in combination with intravenous
immune globulin preparations in transplantation therapy (e.g., bone marrow
transplant).
[0429] In an additional embodiment, the antibodies of the invention are
administered alone or in combination with an anti-inflammatory agent. Anti-
inflammatory agents that may be administered with the antibodies of the
invention include, but are not limited to, glucocorticoids and the
nonsteroidal
anti-inflanunatories, aminoarylcarboxylic acid derivatives, arylacetic acid
derivatives, arylbutyric acid derivatives, arylcarboxylic acids, arylpropionic

acid derivatives, pyrazoles, pyrazolones, salicylic acid derivatives,
thiazinecarboxamides, e-acetarnidocaproic acid, S-adenosylmethionine, 3-
amino-4-hydroxybutyric acid, amixetrine, bendazac, benzydamine, bucolome,
difenpiramide, ditazol, emorfazone, guaiazulene, nabumetone, nimesulide,
orgotein, oxaceprol, paranyline, perisoxal, pifoxime, proquazone, proxazole,
and tenidap.
[0430] In another embodiment, antibodies of the invention are
administered in
combination with a chemotherapeutic agent. Chemotherapeutic agents that
may be administered with the antibodies of the invention include, but are not
limited to, antibiotic derivatives (e.g., doxorubicin, bleomycin,
daunorubicin,

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
159
and dactinomycin); antiestrogens (e.g., tamoxifen); antimetabolites (e.g.,
fluorouracil, 5-FU, methotrexate, floxuridine, interferon alpha-2b, glutamic
acid, plicamycin, mercaptopurine, and 6-thioguanine); cytotoxic agents (e.g.,
carmustine, BCNU, lomustine, CCNU, cytosine arabinoside,
cyclophosphamide, estramustine, hydroxyurea, procarbazine, mitomycin,
busulfan, cis-platin, and vincristine sulfate); hormones (e.g.,
medroxyprogesterone, estramustine phosphate sodium, ethinyl estradiol,
estradiol, megestrol acetate, methyltestosterone, diethylstilbestrol
diphosphate,
chlorotrianisene, and testolactone); nitrogen mustard derivatives (e.g.,
mephalen, chorambucil, mechlorethamine (nitrogen mustard) and thiotepa);
steroids and combinations (e.g., bethamethasone sodium phosphate); and
others (e.g., dicarbazine, asparaginase, mitotane, vincristine sulfate,
vinblastine sulfate, and etoposide).
[0431] In a specific embodiment, antibodies of the invention are
administered
in combination with CHOP (cyclophosphamide, doxorubicin, vincristine, and
prednisone) or any combination of the components of CHOP. In another
embodiment, antibodies of the invention are administered in combination with
Rituximab. In a further embodiment, antibodies of the invention are
administered with Rituxmab arid CHOP, or Rituxmab and any combination of
the components of CHOP.
[0432] In an additional embodiment, the antibodies of the invention are
administered in combination with cytokines. Cytokines that may be
administered with the antibodies of the invention include, but are not limited

to, IL2, IL3, IL4, ILS, I1L6, IL7, IL10, IL12, IL13, IL15, anti-CD40, CD4OL,
IFN-gamma and TNF-alpha. In another embodiment, antibodies of the
invention may be administered with any interleukin, including, but not limited

to, IL-lalpha, IL-lbeta, IL-2, IL-3, IL-4, IL-5, IL-6, M-7, IL-8, M-9, IL-10,
M-
11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, and IL-21.
[0433] In an additional embodiment, the antibodies of the invention are
administered in combination with angiogenic proteins. Angiogenic proteins
that may be administered with the antibodies of the invention include, but are

CA 02548180 2011-12-28
160
not limited to, Glioma Derived Growth Factor (GDGF), as disclosed in
European Patent Number EP-399816; Platelet Derived Growth Factor-A
(PDGF-A), as disclosed in European Patent Number EP-6821 10; Platelet
Derived Growth Factor-B (PDGF-B), as disclosed in European Patent Number
EP-282317; Placental Growth Factor (PIGF), as disclosed in International
Publication Number WO 92/06194; Placental Growth Factor-2 (PIGF-2), as
disclosed in Hauser et al., Gorwth Factors, 4:259-268 (1993); Vascular
Endothelial Growth Factor (VEGF), as disclosed in International Publication
Number WO 90/13649; Vascular Endothelial Growth Factor-A (VEGF-A), as
disclosed in European Patent Number EP-506477; Vascular Endothelial
Growth Factor-2 (VEGF-2), as disclosed in International Publication Number
WO 96/39515; Vascular Endothelial Growth Factor B (VEGF-3); Vascular
Endothelial Growth Factor B-186 (VEGF-B186), as disclosed in International
Publication Number WO 96/26736; Vascular Endothelial Growth Factor-D
(VEGF-D), as disclosed in International Publication Number WO 98/02543;
Vascular Endothelial Growth Factor-D (VEGF-D), as disclosed in
International Publication Number WO 98/07832; and Vascular Endothelial
Growth Factor-E (VEGF-E), as disclosed in German Patent Number
DE19639601.
[0434] In an additional embodiment, the antibodies of the invention are
administered in combination with hematopoietic growth factors.
Hematopoietic growth factors that may be administered with the antibodies of
TM
the invention include, but are not limited to, LEUKINE
TM TM TM
(SARGRAMOSTIM ) and NEUPOGEN (FILGRASTIM ).
[0435] In an additional embodiment, the antibodies of the invention are
administered in combination with Fibroblast Growth Factors. Fibroblast
Growth Factors that may be administered with the antibodies of the invention
include, but are not limited to, FGF-1, FGF-2, FGF-3, FGF-4, FG17-5, FGF-6,
FGF-7, FGF-8, FGF-9, FGF-10, FGF-11, FGF-12, FGF-13, FGF-14, and FGF-
15.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
161
EXAMPLE 1
C35 EXPOSED ON SURFACE MEMBRANE OF BREAST TUMOR CELLS
FOLLOWING RADIATION INDUCED APOPTOSIS
[0436] A line of continuously growing breast tumor cells that express the
C35
tumor antigen was either irradiated with 300 Gy or left untreated. After
continued in vitro culture for several days to allow apoptosis to develop,
cells
were harvested, washed and stained with 50 ng of 1F2 monoclonal anti-C35
antibody or a mouse IgG antibody control each conjugated to the fluorescent
dye Alexa 647. Following 50 minutes incubation at 25C, cells were stained
with Annexin V and propidium iodide (PI) using a standard commercial kit
(Pharmingen). Cells were analyzed for staining with Annexin V, propidium
iodide and Alexa 647 by flow cytometry employing standard protocols.
[0437] The results in Figure 1 show that untreated live cells (PI
negative), that
are not undergoing apoptosis (Annexin V negative), do not express C35 on the
surface membrane as evidenced by absence of differential staining with anti-
C35 antibody and the isotype control antibody (Figure 1A). Similarly,
irradiated tumor cells that remain viable (PI negative) and have not been
induced to undergo apoptosis (Annexin V negative) also do not express C35
on the tumor cell surface membrane (Figure 1B). In striking contrast,
irradiated tumor cells that are viable (PI negative), but undergoing apoptosis

(Annexin V positive), are clearly differentially stained with anti-C35
antibodies as compared to isotype control antibody (Figure 1C).
EXAMPLE 2
C35 EXPOSED ON SURFACE MEMBRANE OF BREAST TUMOR CELLS
FOLLOWING DRUG INDUCED APOPTOSIS
[0438] A line of continuously growing breast tumor cells that express the
C35
tumor antigen was either treated with 6ug/m1 mitomycin C or left untreated.
After continued in vitro culture for 48 hours to allow apoptosis to develop,

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
162
cells were harvested, washed and stained with 50 ng of 1F2 monoclonal anti-
C35 antibody or a mouse IgG antibody control each conjugated to the
fluorescent dye Alexa 647. Following 50 minutes incubation at 25C, cells
were stained with Annexin V and propidium iodide (PI) using a standard
commercial kit (Pharmingen). Cells were analyzed for staining with Annexin
V, propidium iodide and Alexa 647 by flow cytometry employing standard
protocols.
[0439] The results in Figure 2 show that untreated live cells (PI
negative), that
are not undergoing apoptosis (Annexin V negative), do not express C35 on the
surface membrane as evidenced by absence of differential staining with anti-
C35 antibody and the isotype control antibody (Figure 2A). Similarly,
mitomycin C treated tumor cells that remain viable (PI negative) and have not
been induced to undergo apoptosis (Annexin V negative) also do not express
C35 on the tumor cell surface membrane (Figure 2B). In striking contrast,
mitomycin C treated tumor cells that are viable (PI negative), but undergoing
apoptosis (Annexin V positive), are clearly differentially stained with anti-
C35
antibodies as compared to isotype control antibody (Figure 2C).
EXAMPLE 3
EXPRESSION OF AN ANTIBODY IN MAMMALIAN CELLS
[0440] The polypeptide of the present invention can be expressed in a
mammalian cell. A typical mammalian expression vector contains a promoter
element, which mediates the initiation of transcription of mRNA, a protein
coding sequence, and signals required for the termination of transcription and

polyadenylation of the transcript. Additional elements include enhancers,
Kozak sequences and intervening sequences flanked by donor and acceptor
sites for RNA splicing. Highly efficient transcription is achieved with the
early
and late promoters from SV40, the long terminal repeats (LTRs) from
Retroviruses, e.g., RSV, HTLVI, HIVI and the early promoter of the

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
163
cytomegalovirus (CMV). However, cellular elements can also be used (e.g.,
the human actin promoter).
[0441] Suitable expression vectors for use in practicing the present
invention
include, for example, vectors such as pSVL and pMSG (Pharmacia, Uppsala,
Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146), pBC12MI
(ATCC 67109), pCMVSport 2.0, and pCMVSport 3Ø Mammalian host cells
that could be used include, human Hela, 293, H9 and Jurkat cells, mouse
NIH3T3 and C127 cells, Cos 1, Cos 7 and CV1, quail QC1-3 cells, mouse L
cells and Chinese hamster ovary (CHO) cells.
[0442] Alternatively, the polyp eptide can be expressed in stable cell
lines
containing the polynucleotide integrated into a chromosome. The co-
transfection with a selectable marker such as DHFR, gpt, neomycin,
hygromycin allows the identification and isolation of the transfected cells.
[0443] The transfected gene can also be amplified to express large
amounts of
the encoded protein. The DHFR (dihydrofolate reductase) marker is useful in
developing cell lines that carry several hundred or even several thousand
copies of the gene of interest. (See, e.g., Alt, F. W., et al., J. Biol. Chem.

253:1357-1370 (1978); Hamlin, J. L. and Ma, C., Biochem. et Biophys. Acta,
1097:107-143 (1990); Page, M. J. and Sydenham, M. A., Biotechnology 9:64-
68 (1991).) Another useful selection marker is the enzyme glutamine synthase
(GS) (Murphy et al., Biochem J. 227:277-279 (1991); Bebbington et al.,
Bio/Technology 10:169-175 (1992). Using these markers, the mammalian
cells are grown in selective medium and the cells with the highest resistance
are selected. These cell lines contain the amplified gene(s) integrated into a

chromosome. Chinese hamster ovary (CHO) and NSO cells are often used for
the production of proteins.
[0444] Derivatives of the plasmid pSV2-dhfr (ATCC Accession No. 37146),
the expression vectors pC4 (ATCC Accession No. 209646) and pC6 (ATCC
Accession No.209647) contain the strong promoter (LTR) of the Rous
Sarcoma Virus (Cullen et al., Molecular and Cellular Biology, 438-447
(March, 1985)) plus a fragment of the CMV-enhancer (Boshart et al., Cell

CA 02548180 2011-12-28
164
41:521-530 (1985).) Multiple cloning sites, e.g., with the restriction enzyme
cleavage sites BamHI, XbaI and Asp718, facilitate the cloning of the gene of
interest. The vectors also contain the 3' intron, the polyadenylation and
termination signal of the rat preproinsulin gene, and the mouse DHFR gene
under control of the SV40 early promoter.
[0445] Specifically, the plasmid pC6, for example, is digested with
appropriate restriction enzymes and then dephosphorylated using calf
intestinal
phosphates by procedures known in the art. The vector is then isolated from a
1% agarose gel.
[0446] A polynucleotide of the present invention is amplified according to
protocols known in the art. If a naturally occurring signal sequence is used
to
produce the polypeptide of the present invention, the vector does not need a
second signal peptide. Alternatively, if a naturally occurring signal sequence
is
not used, the vector can be modified to include a heterologous signal
sequence.
(See, e.g., WO 96/34891.)
[0447] The amplified fragment is isolated from a 1% agarose gel using a
TM
commercially available kit ("Geneclean," BIO 101 Inc., La Jolla, Calif.). The
fragment then is digested with appropriate restriction enzymes and again
purified on a 1% agarose gel.
104481 The amplified fragment is then digested with the same restriction
enzyme and purified on a 1% agarose gel. The isolated fragment and the
dephosphorylated vector are then ligated with T4 DNA ligase. E. coli HB101
or XL-1 Blug-Cells are then transformed and bacteria are identified that
contain
the fragment inserted into plasmid pC6 using, for instance, restriction enzyme

analysis.
[0449] Chinese hamster ovary cells lacking an active DHFR gene is used for
transfection. Five µg of the expression plasmid pC6 or pC4 is cotransfected

with 0.5 µg of the plasmid pSVneo using lipofectiiim(Felgner et al.,
supra).
The plasmid pSV2-neo contains a dominant selectable marker, the neo gene
from Tn5 encoding an enzyme that confers resistance to a group of antibiotics
including G418. The cells are seeded in alpha minus MEM supplemented with

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
165
1 mg/ml G418. After 2 days, the cells are trypsinized and seeded in hybridoma
cloning plates (Greiner, Germany) in alpha minus MEM supplemented with
10, 25, or 50 ng/ml of metothrexate plus 1 mg/ml G418. After about 10-14
days single clones are trypsinized and then seeded in 6-well petri dishes or
10
ml flasks using different concentrations of methotrexate (50 nM, 100 nM, 200
nM, 400 nM, 800 nM). Clones growing at the highest concentrations of
methotrexate are then transferred to new 6-well plates containing even higher
concentrations of methotrexate (1 uM, 2 uM, 5 uM, 10 mM, 20 mM). The
same procedure is repeated until clones are obtained which grow at a
concentration of 100-200 uM. Expression of the desired gene product is
analyzed, for instance, by SDS-PAGE and Western blot or by reversed phase
HPLC analysis.
EXAMPLE 4
RADIOLABELED C35-SPECIFIC ANTIBODIES CONCENTRATE IN
NECROTIC REGIONS OF VIABLE TUMORS EXPRESSING C35.
[0450] BALB/c mice were engrafted on opposite flanks with syngeneic non-
small cell lung cancer derived Line 1 tumor cells that either had or had not
been transfected with human C35. C35 protein expression was confirmed by
immunohistochemical staining with anti-C35 antibodies. After 14 days in
vivo growth, animals received intravenous injection of 125I-labeled anti-C35
antibody. Animals were sacrificed 120 hrs after injection of radiolabeled
antibodies and the concentration of anti-C35 antibodies in C35-positive and
C35-negative tumors was determined by exposure of a tumor section to film.
As shown in Figure 4, radiolabeled anti-C35 antibodies concentrated only in
the C35-positive and not the C35-negative tumors. Comparison of the
distribution of label and an H&E stain for intact cells within the tumors,
confirmed that under these conditions the labeled anti-C35 antibodies
concentrated specifically in the necrotic regions of the C35-positive tumor.

CA 02548180 2011-12-28
166
EXAMPLE 5
PROTOCOL FOR ADMINISTRATION OF DOT-METRIC AND
THERAPEUTIC RADIOLABELED ANTIBODY
[0451] The radiolabeled
antibody (or antibody fragment) compositions, which
include both the dosimetric radiolabeled antibody and the therapeutic
radiolabeled antibody, are administered intravenously or intraarterially in
the
form of an injection. The injectable radiolabeled antibody compositions will
be infused into a vein or artery over the course of 5 minutes to about 60
minutes, preferably from 15 minutes to 30 minutes. Where the tumor is
supplied by a known artery, intraarterial administration is preferred for the
therapeutic radiolabeled antibody compositions. Both the
dosimetric
radiolabeled antibody and the therapeutic radiolabeled antibody will be
administered as sterile aqueous solutions typically in physiologic phosphate-
buffered saline or other vehicle suitable for parenteral injection. The
initial
dosimetric radiolabeled antibody dose will be approximately 5-100mg of
antibody which will deliver approximately 5-50mCi radiation. Approximately
5-10 days following the dosimetric dose, the therapeutic radiolabeled antibody

will be administered at a dose of approximately 10-500 mg which will deliver
as much as 300 mCi radiation for each therapeutic dose. This
dosimetric/therapeutic regimen may be repeated. See also, US 5,057,313 and
US 5,120,525.
EXAMPLE 6
CLONING OF ANTI-C35 MOUSE AND HUMAN ANTIBODY VARIABLE
REGION GENES INTO DEPOSITED TOPO CLONES
[0452] The
immunoglobulin heavy and light chain variable regions were
TM -rm
cloned into the TOPO vector (Invitrogen) by PCR amplification of the V
region and TA cloning into the TOPOmvector. This ligation system does not
TM
require restriction enzyme digestion (although the TOPO vector does

CA 02548180 2011-12-28
167
incorporate EcoRI sites to allow subsequent excision of inserts). TA cloning
takes advantage of naturally added 3' A overhangs in the PCR amplification
product of Taq polymerase which can then pair with 5' T overhangs in the
linearized vector provided in the TOPO cloning kit (Invitrogen).
[0453] To PCR amplify variable region genes for insertion into TOPO, we
employed a downstream primer complementary to the 5' end of the constant
region sequence (different for heavy and light chains and for mouse and
human) and a known fixed primer sequence added at the 5' end of the variable
region by 5' RACE using the Invitrogen GeneRacer kit. These methods are
well known to those skilled in the art.
EXAMPLE 7
CLONING VARIABLE GENES FROM DEPOSITED TOPO CLONES INTO
pCMV EXPRESSION CONSTRUCTS
Generation of pCMV expression constructs
[0454] The construction of vaccinia transfer plasmids ¨ pVHE, pVKE and
pVLE ¨ has been described in a previous patent application (US 2002 0123057
Al, "In vitro Methods of Producing and Identifying Imsnunoglobulin
Molecules in Eukaryotic Cells", published 2002-09-05). To generate the
mammalian expression vectors to express the immunoglobulin heavy and light
chains, the expression cassettes, from NotI to Sall, were excised from these
vaccinia transfer plasmids and cloned into the pCMV-Script vector (whose
XhoI site in the vector multiple cloning site was destroyed by fill-in and
blunt
end ligation), resulting in the generation of pCMV-VH, pCMV-VK and
pCMV-VL vectors. These expression cassettes contain the signal peptide,
cloning sites for the V genes and the constant regions from the membrane-
bound p. heavy chain and the K light chain genes.
[0455] In pCMV-VH, the cassette contains the signal peptide from amino acid
position -19 relative to the start codon [aa(-19)] to aa(-3), followed by
aa(109

CA 02548180 2011-12-28
168
to 113) of the VH genes and the whole heavy chain constant region. The
selected VH genes, from aa(-4) to aa(110) can be cloned into pCMV-VH at
BssHIE [aa(-4 to -3)] and BstEll [aa(109-110)] sites.
[0456] In pCMV-VK (kappa), the cassette contains the signal peptide
from
aa(-19) to aa(-2), followed by aa(104 to 107) of the VK genes and the whole
kappa chain constant region. The selected VK genes, from aa(-3) to aa(105)
can be cloned into pCMV-VK at ApaLI [aa(-3 to -2)] and XhoI [aa(104-105)]
sites.
[0457] In pCMV-VL (lambda), the cassette contains the signal peptide
from
aa(-19) to aa(-2), followed by aa(103 to 107) of the VL genes and the whole
kappa chain constant region. The selected VL genes, from aa(-3) to aa(104)
can be cloned into pCMV-VL at ApaLI [aa(-3 to -2)] and Hindi-IT [aa(103-
104)] sites. The resulting lambda light chain will exhibit the VkCI( chimeric
structure.
[0458] To express the selected antibodies as secreted human IgGl, the
constant region of IgG1 was cloned from B cells or bone marrow cells by RT-
PCR. The primer set used was:
5' forward primer: 5'-ATTAGGATCCGGTCACCGTCTCCTCAGCC-3'
(SEQ ID NO:15)
3' reverse primer: 5'-ATTAGTCGACTCATTTACCCGGAGACAGGGA-3'
(SEQ ID NO:16)
[0459] The resulting PCR
product exhibits the following structure: BamIll-
BstEll(aal09-110)-(aa111-113)-Cy1-TGA-SalL The PCR product
was
TM
subcloned into pBluescriptIVKS at BamHI and Sall sites to carry out site
directed mutagenesis employing standard protocols to remove the internal
BstEll located at the CHI region via silent mutation. The resulting Cyi was
then subcloned into pCMV-VH at BstEll and Sall to generate pCMV-Cyi to
direct the expression of secreted IgG1 heavy chain, once a VH gene is
subcloned into this vector at BssIIII/BstEn.
[0460] The sequence of IgGl-secreted, human gammal heavy chain leader
and constant region cassette for insertion of V genes follows.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
169
Underline = restriction sites
Bold = Constant region
Bold/italics = Signal peptide
Not 1 NcoI
goggccgcaaaccatgggatggagctgtatcatcctottettggtagcaacagetacag
BssHII BsteII
gcgcgoatatggtcaccgtotcctcagectecaccaagggcccateggtetteccectggcac
cc tcctccaagagcacc tc tgggggcacageggccctgggctgcc tggtcaaggactacttcc
ccgaacaggtgacggtgtcgtggaactcaggcgccctgaccageggcgtgcacaccttcccgg
ctgtectacagtcctcaggactctactccctcagcagcgtegtgaccgtgccctccagcagct
tgggcacccagacc tacatctgcaacgtgaatcacaagcccagcaacaccaaggtggacaaga
aagttgagcccaaatc t tgtgacaaaac tcacacatgaccaccgtgcccagcacctgaactcc
tggggggaccgtcagtettcctettccecccaaaacccaaggacaccetcatgatcteccgga
ccectgaggtcacatgegtggtggtggacgtgagccacgaagaccc tgaggtcaagttcaac t
ggtacgtggacggcgtggaggtgcataatgccaagacaaagccgcgggaggagcagtacaaca
gcacgtaccgtgtggtcagcgtcatcaccgtectgcaccaggac tggc tgaatggcaaggagt
acaagtgcaaggtctccaacaaagccetcccagcccccatcgagaaaaccatctccaaagcca
aagggcagccccgagaaccacaggtgtacaccctgcccccatcccgggatgagctgaccaaga
accaggtcagcctgacctgcctggecaaaggcttctatcccagcgacatcgccgtggagtggg
agagcaatgggcagccggagaacaactacaagaccacgccteccgtgctg-gactccgacgget
cc t tcttcc tctacagcaagetcaccgtggacaagagcaggtggcagcaggggaacgtcttc t
catgctccgtgatgcatgaggc tctgcacaaccac tacacgcagaagagcc tctccct
Sall
Gtctccgggtaaatgagtcgac (SEQ ID NO:17)
[0461] The sequence of human light chain leader and kappa constant region
cassette for insertion of Vic genes follows.
Not 1 NcoI
goggccgcaaaccatgggatggagctgtatcatectottottggtagcaacagctacag
ApaLl XhoI
gcgtgcacttgactcgagatcaaacgaactgtggctgcacca totgtottcatettoccgcca
tetgatgagcagttgaaatctggaactgcctctgttgtgtgcctgctgaataacttctatccc
agagaggccaaagtacagtggaaggtggataacgccc tccaa tcgggtaac tcccaggagagt
gtcacagagcaggacagcaaggacagcacctacagcc tcagcagcaccctgacgctgagcaaa
gcagactacgagaaacacaaagtctacgcctgcgaagtcaccc
Sall
Atcagggcctgagc tcgcccgtcacaaagagettcaacaggggagagtgttaggtcgac
(SEQ ID NO:18)

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
170
[0462] The sequence of human light chain leader and kappa constant region
cassette for insertion of Vk genes follows.
Not 1 Ncol
gcggccgcaaaccatgggatggagctgtatcatcctettcttggtagcaacagctacag
ApaLl HindIII
gcgtgeacttgactcgagaagcttaccgtcctacgaactgtggetgcaccatctgtettcato
ttcccgccatctgatgagcagttgaaatctggaactgcctctgttgtgtgcctgctgaataac
ttctatcccagagaggccaaagtacagtggaaggeggataacgccctccaatcgggtaactcc
caggagagtgtcacagagcaggacagcaaggacagcacctacagcctcagcagcaccctgacg
ctgagcaaagcagactacgagaaacacaaagtctacgcctgcgaagtcacccatcagggcctg
agctcgcccgtcacaaagagcttcaacaggggagagtgt
SAL 1
Taggtcgac (SEQ ID NO:19)
[0463] To construct vectors that express secreted human antibodies of
other
isotypes, including secreted forms of IgG2, IgG3, IgG4, IgA, IgD, IgE and
IgM, the same approach can be taken to clone the respective constant regions,
to mutagenize any internal BstEII site, and to substitute the Gyi with the
constant regions of other isotypes between the BstEII and Sall sites in the
pCMV-Cyi vector.
[0464] To clone the constant regions of other isotypes, the following
primer
pairs were used:
IgG2-F: 5'-ATTAGGATCCGGTCACCGTCTCCTCAGCC-3' (SEQ ID
NO:20)
IgG2-R: 5'-ATTAGTCGACTCATTTACCCGGAGACAGGGA-3' (SEQ ID
NO :21)
IgG3-F: 5'-ATTAGGATCCGGTCACCGTCTCCTCAGCT-3' (SEQ ID
NO:22)
IgG3-R: 5'-ATTAGTCGACTCATTTACCCGGAGACAGGGA-3' (SEQ ID
NO:23)
IgG4-F: 5'-ATTAGGATCCGGTCACCGTCTCCTCAGCT-3' (SEQ ID
NO:24)
IgG4-R: 5'-ATTAGTCGACTCATTTACCCAGAGACAGGGA-3' (SEQ ID
NO:25)

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
171
IgAl-F: 5' -ATTAGGATCCGGTCACCGTCTCCTCAGCAT-3' (SEQ ID
NO:26)
IgAl-R: 5'-ATTAGTCGACTCAGTAGCAGGTGCCGTCCAC-3' (SEQ ID
NO:27)
IgA2-F: 5'-ATTAGGATCCGGTCACCGTCTCCTCAGCAT-3' (SEQ ID
NO:28)
IgA2-R: 5'-ATTAGTCGACTCAGTAGCAGGTGCCGTCCAC-3' (SEQ ID
NO:29)
IgD-F: 5' -ATTAGGATCCGGTCACCGTCTCCTCAGCAC-3' (SEQ ID
NO:30)
IgD-R: 5'-ATTAGTCGACTCATTTCATGGGGCCATGGTC-3' (SEQ ID
NO :31)
IgE-F: 5' -ATTAGGATCCGGTCACCGTCTCCTCAGCC-3' (SEQ ID
NO:32)
IgE-R: 5'-ATTAGTCGACTCATTTACCGGGATTTACAGA-3' (SEQ ID
NO:33)
IgM-F: 5'-ATTAGGATCCGGTCACCGTCTCCTCAGGG-3' (SEQ ID
NO:34)
IgM-R: 5'-ATTAGTCGACTCAGTAGCAGGTGCCAGCTGT-3' (SEQ ID
NO :35)
[0465] Note that, because of the high degree of sequence conservation,
primers used are the same between IgG1 and IgG2, between IgG3 and IgG4,
and between IgAl and IgA2.
Cloning Variable genes from Topo clones into pCMV expression constructs
[0466] Step 1: Generation of V-gene fragments.
A. Human v-genes
MMH1

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
172
1. Digest MMH1 plasmid DNA (clone H0009) with
BssHII (GCGCGC(SEQ ID NO:36)) and Bstell (GGTCACC (SEQ ID
NO:37)) using standard protocols.
2. Resolve DNA on agarose gel using standard protocols.
3. Excise 357 bp fragment from gel and isolate DNA using
standard protocols.
MMK1
1. Digest MMK1 plasmid DNA (clone L0010) with ApaLl
(GTGCAC(SEQ ID NO:38)) and Xhol (CTCGAG (SEQ ID NO:39)) using
standard protocols.
2. Resolve DNA on agarose gel using standard protocols.
3. Excise 343 bp fragment from gel and isolate DNA using
standard protocols.
B. Mouse hybridoma v-genes:
1F2VK
1. The mouse hybridoma v-gene must be PCR amplified
from the ATCC deposited clone 1F2K using the following primers. This is
necessary to create a chimeric antibody of the mouse v-gene with human
constant region in the human kappa light chain constant region expression
cassette.
1F2VK forward primer:
5' ¨ tatccgtgcactccCAAATTGTTCTCACCCAGTCTCCAG ¨ 3' (SEQ ID
NO:40)
1F2VK reverse primer:
5' ¨ atattctcgAGCTTGGTCCCCCCTCCGAA ¨3' (SEQ ID NO:41)
(Lowercase = non-homologous to mouse 1F2 VK sequence, includes
restriction site. CAPITALS= homologous to mouse 1F2 VK sequence)
2. Digest 331 bp PCR product with ApaL1 (GTGCAC
(SEQ ID NO:42)) and Xhol (CTCGAG (SEQ ID NO:43)) using standard
protocols.
3. Resolve DNA on agarose gel using standard protocols.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
173
4. Excise 315 bp digested fragment from gel and isolate
DNA using standard protocols.
1F2VH
1. The mouse hybridoma v-gene must be PCR amplified
from the ATCC deposited clone 1F2G using the following primers. This is
necessary to create a chimeric antibody of the mouse v-gene with human
constant region in the human heavy chain constant region expression cassette.
1F2VH forward primer:
5' ¨ tataagcgcgcactccGATGTACAGCTTCAGGAGTCAGGAC (SEQ ID
NO:44)
1F2VH reverse primer:
5'-atattgGTGACCAGAGTCCCTTGGCCCC-3' (SEQ ID NO:45)
(Lowercase = non-homologous, contains restriction sites. CAPITALS=
homologous)
2. Digest 360 bp PCR product with BssHII (GCGCGC
(SEQ ID NO:36)) and Bstell (GGTCACC (SEQ ID NO:37)) using standard
protocols.
3. Resolve DNA on agarose gel using standard protocols.
4. Excise gel slice containing 343 bp digested DNA
fragment and isolate DNA using standard protocols.
1B3VK
1. The mouse hybridoma v-gene must be per amplified
from the deposited clone 1B3K using the following primers. This is necessary
to create a chimeric antibody of the mouse v-gene with human constant region
in the human kappa light chain constant region expression cassette.
1B3VK forward primer:
5' ¨ tatccgtgcactccGATGTCCAGATAACCCAGTCTCCATC ¨ 3' (SEQ ID
NO:46)
1B3VK reverse primer:
5' ¨atattctcgAGCTTGGTCCCAGCACCGAA ¨3' (SEQ ID NO:47)

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
174
(Lowercase = non-homologous, contains restriction sites. CAPITALS=
homologous)
2. Digest 334 bp PCR product with ApaLl (GTGCAC
(SEQ ID NO:42)) and Xhol (CTCGAG (SEQ ID NO:43)) using standard
protocols.
3. Resolve DNA on agarose gel using standard protocols.
4. Excise gel slice containing digested 322 bp DNA
fragment and isolate DNA using standard protocols
1B3VH
1. The mouse hybridoma v-gene must be per amplified
from the deposited clone 1B3G using the following primers. This is necessary
to create a chimeric antibody of the mouse v-gene with human constant region
in the human heavy chain constant region expression cassette.
1B3VH forward primer:
5'- tataagcgcgcactccGAGGTGCAGCTTCAGGAGTCAGGAC ¨3' (SEQ ID
NO:48)
1B3VH reverse primer:
5' ¨atattGGTGACCGTGGTCCCAGCG ¨3' (SEQ ID NO:49)
(Lowercase = non¨homologous, contains restriction sites. CAPITALS=
homologous
2. Digest 378 bp PCR product with BssHII (GCGCGC
(SEQ lD NO:36)) and Bstell (GGTCACC (SEQ ID NO:37)) using standard
protocols.
3. Resolve DNA on agarose gel using standard protocols.
4. Excise gel slice containing 366 bp digested DNA
fragment and isolate DNA using standard protocols.
[0467] Step 2: Assembly of expression constructs
1. Digest pCMV-VH and pCMV-VK expression vectors with the
appropriate enzymes using standard protocols:
a. pCMV-VH ¨ Bsten and BssHII
b. pCMV-VK¨ ApaLl and Xho I

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
175
2. Resolve DNA on agarose gel and excise linearized vector using
standard protocols.
3. Isolate DNA from gel slice using standard protocols.
4. Ligate light chain v-genes into pCMV-VK and heavy chain v-
genes into pCMV-VH using standard protocols.
5. Transform ligated DNA into competent cells and isolate
plasmid DNA using standard protocols.
EXAMPLE 8
SEQUENCES OF IMMUNOGLOBULINT CONSTANT REGIONS
[0468] The following genes and encoded amino acids sequences may be used
to prepare humanized antibodies, human variant antibodies, chimeric
antibodies, and fragments thereof.
[0469] Homo sapiens G2 gene for irnmunoglobulin constant region (IgG2 (n-)
allotype) (GenBank No. Z49802) (SEQ ID NO:50)
1 tcttctctct gcagagcgca aatgttgtgt cgagtgccca ccgtgcccag gtaagccagc
61 ccaggcctcg ccctccagct caaggcggga caggtgccct agagtagcct gcatccaggg
121 acaggcccca gctgggtgct gacacgtcca cctccatctc ttcctcagca ccacctgtgg
181 caggaccgtc agtcttcctc ttccccccaa aacccaagga caccctcatg atctcccgga
241 ccectgaggt cacgtgcgtg gtggtggacg tgagccacga agaccccgag gtccagttca
301 actggtacgt ggacggcgtg gaggtgcata atgccaagac aaagccacgg gaggagcagt
361 tcaacagcac gttccgtgtg gtcagcgtcc tcaccgttgt gcaccaggac tggctgaacg
421 gcaaggagta caagtgcaag gtctccaaca aaggcctccc agcccccatc gagaaaacca
481 tctccaaaac caaaggtggg acccgcgggg tatgagggcc acatggacag acggcggctt
541 cggcccaccc tctgccctgg gagtgaccgc tgtgccaacc tctgtcccta cagggcagcc
601 ccgagaacca caggtgtaca ccctgccccc atcccgggag gagatgacca agaaccaggt
661 cagcctgacc tgcctggtca aaggcttcta ccccagcgac atcgccgtgg agtgggagag
721 caatgggcag ccggagaaca actacaagac cacacctccc atgctggact ccgacggctc

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
176
781 cttcttcctc tacagcaagc tcaccgtgga caagagcagg tggcagcagg ggaacgtctt
841 ctcatgctcc gtgatgcatg aggctctgca caaccactac acgcagaaga gcctctccct
901 gtctccgggt aaatgagtgc cacggccggc aagcc
[0470] H. sapiens G2 gene for immunoglobulin constant region (IgG2 (n+)
allotype) (GenBank No. Z49801) (SEQ ID NO:51)
1 tcttctctct gcagagcgca aatgttgtgt cgagtgccca ccgtgcccag gtaagccagc
61 ccaggcctcg ccctccagct caaggcggga caggtgccct agagtagcct gcatccaggg
121 acaggcccca gctgggtgct gacacgtcca cctccatctc ttcctcagca ccacctgtgg
181 caggaccgtc agtcttcctc ttccccccaa aacccaagga caccctcatg atctcccgga
241 cccctgaggt cacgtgcgtg gtggtggacg tgagccacga agaccccgag gtccagttca
301 actggtacgt ggacggcatg gaggtgcata atgccaagac aaagccacgg gaggagcagt
361 tcaacagcac gttccgtgtg gtcagcgtcc tcaccgtcgt gcaccaggac tggctgaacg
421 gcaaggagta caagtgcaag gtctccaaca aaggcctccc agcccccatc gagaaaacca
481 tctccaaaac caaaggtggg acccgcgggg tatgagggcc acatggacag acggcggctt
541 cggcccaccc tctgccctgg gagtgaccgc tgtgccaacc tctgtcccta cagggcagcc
601 ccgagaacca caggtgtaca cectgccccc atcccgggag gagatgacca agaaccaggt
661 cagcctgacc tgcctggtca aaggcttcta ccccagcgac atcgccgtgg agtgggagag
721 caatgggcag ccggagaaca actacaagac cacacctccc atgctggact ccgacggctc
781 cttcttcctc tacagcaagc tcaccgtgga caagagcagg tggcagcagg ggaacgtctt
841 ctcatgctcc gtgatgcatg aggctctgca caaccactac acacagaaga gcctctccct
901 gtctccgggt aaatgagtgc cacggccggc aagcc
[0471] Homo sapiens CH gene encoding immunoglobulin, constant region,
heavy chain, alpha-2 subunit (GenBank No. AJ012264) (SEQ ID NO:52)
1 ctcgaggacc tgctcttagg ttcagaagcg aacctcacgt gcacactgac cggcctgaga
61 gatgcctctg gtgccacctt cacctggacg ccctcaagtg ggaagagcgc tgttcaagga
121 ccacctgagc gtgacctctg tggctgctac agcgtgtcca gtgtcctgcc tggctgtgcc
181 cagccatgga accatgggga gaccttcacc tgcactgctg cccaccccga gttgaagacc
241 ccactaaccg ccaacatcac aaaatccggt gggtccagac cctgctcggg gccctgctca
301 gtgctctggt ttgcaaagca tattcctggc ctgcctcctc cctcccaatc ctgggctcca
361 gtgctcatgc caagtacaca gggaaactga ggcaggctga ggggccagga cacagcccag

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
177
421 ggtgcccacc agagcagagg ggctctctca tcccctgccc agccccctga cctggctctc
481 taccctccag gaaacacatt ccggcccgag gtccacctgc tgccgccgcc gtcggaggag
541 ctggccctga acgagctggt gacgctgacg tgcctggcac gtggcttcag ccccaaggat
601 gtgctggttc gctggctgca ggggtcacag gagctgcccc gcgagaagta cctgacttgg
661 gcatcccggc aggagcccag ccagggcacc accacctacg ctgtaaccag catactgcgc
721 gtggcagctg aggactggaa gaagggggag accttctcct gcatggtggg ccacgaggcc
781 ctgccgctgg ccttcacaca gaagaccatc gaccgcatgg cgggtaaacc cacccacatc
841 aatgtgtctg ttgtcatggc ggaggcggat ggcacctgct actgagccgc ccgcctgtcc
901 ccacccctga ataaactcca tgctccccca agcagcccca cgcttccatc cggcgcctgt
961 ctgtccatcc tcagggtctc agcacttggg aaagggccag ggcatggaca gggaagaata
1021 ccccctgccc tgagcctcgg ggggcccctg gcacccccat gagactttcc accctggtgt
1081 gagtgtgagt tgtgagtgtg agagtgtgtg gtgcaggagg cctcgctggt gtgagatctt
1141 aggtctgcca aggcaggcac agcccaggat gggttctgag agacgcacat gccccggaca
1201 gttctgagtg agcagtggca tggccgtttg tccctgagag agccgcctct ggctgtagct
1261gggagggaat agggagggta aaaggagcag gctagccaag aaaggcgcag gtagtggcag
1321 gagtggcgag ggagtgaggg gctggactcc agggccccac tgggaggaca agctccagga
1381 gggccccacc accctagtgg gtgggcctca ggacgtccca ctgacgcatg caggaagggg
1441 cacctcccct taaccacact gctctgtacg gggcacgtgg gcacacatgc acactcacac
1501 tcacatatac gcctgagccc tgcaggagtg gaacgttcac agcccagacc cagttccaga
1561 aaagccaggg gagtcccctc ccaagccccc aagctcagcc tgctccccca ggcccctctg
1621 gcttccctgt gtttccactg tgcacagctc agggaccaac tccacagacc cctcccaggc
1681 agcccctgct ccctgcctgg ccaagtctcc catcccttcc taagcccaac taggacccaa
1741 agcatagaca gggaggggcc gcgtggggtg gcatcagaag
[0472] Homo sapiens constant region, heavy chain, alpha-2 subunit (GenBank
No. CAA09968.1) (SEQ ID NO:53)
LEDLLLGSEANLTCTLTGLRDASGATFTWTPSSGKSAVQGPPER
DLCGCYSVSSVLPGCAQPWNHGETFTCTAAHPELKTPLTANITKSGNT
FRPEVHLLPPPSEELALNELVTLTCLARGFSPKDVLVRWLQGSQELPRE
KYLTWASRQEPSQGTTTYAVTSILRVAAEDWKKGETFSCMVGHEALP
LAFTQKTIDRMAGKPTHINVSVVMAEADGTCY

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
178
[0473] Homo sapiens partial mRNA for immunoglobulin heavy chain constant
region alpha 1 (IGHAl gene) (GenBank No. AJ294729) (SEQ ID NO:54)
1 gcaagcttga ccagccccaa ggtcttcccg ctgagcctct gcagcaccca gccagatggg
61 aacgtggtca tcgcctgcct ggtccagggc ttctteccce aggagccact cagtgtgacc
121 tggagcgaaa gcggacaggg cgtgaccgcc agaaacttcc cacccagcca ggatgcctcc
181 ggggacctgt acaccacgag cagccagctg accctgccgg ccacacagtg cctagccggc
241 aagtccgtga catgccacgt gaagcactac acgaatccca gccaggatgt gactgtgccc
301 tgcccagttc cctcaactcc acctacccca tetccetcaa ctccacctac cccatctccc
361 tcatgctgcc acccccgact gtcactgcac cgaccggccc tcgaggacct gctcttaggt
421 tcagaagcga acctcacgtg cacactgacc ggcctgagag atgcctcagg tgtcaccttc
481 acctggacgc cctcaagtgg gaagagcgct gttcaaggac cacctgaccg tgacctctgt
541 ggctgctaca gcgtgtccag tgtcctgtcg ggctgtgccg agccatggaa ccatgggaag
601 accttcactt gcactgctgc ctaccccgag tccaagaccc cgctaaccgc caccctctca
661 aaatccggaa acacattccg gcccgaggtc cacctgctgc cgccgccgtc ggaggagctg
721 gccctgaacg agctggtgac gctgacgtgc ctggcacgtg gcttcagccc caaggatgtg
781 ctggttcgct ggctgcaggg gtcacaggag ctgccccgcg agaagtacct gacttgggca
841 tcccggcagg agcccagcca gggcaccacc accttcgctg tgaccagcat actgcgcgtg
901 gcagccgagg actggaagaa gggggacacc ttctcctgca tggtgggcca cgaggccctg
961 ccgctggcct tcacacagaa gaccatcgac cgcttggcgg gtaaacccac ccatgtcaat
1021 gtgtctgttg tcatggcgga ggtggacggc acctgctac
[0474] Imunoglobulin heavy chain constant region alpha 1 (GenBank No.
CAC20453.1) (SEQ ID NO:55)
ASLTSPKVFPLSLCSTQPDGNVVIACLVQGFFPQEPLSVTWSESGQGVT
ARNFPPSQDASGDLYTTSSQLTLPATQCLAGKSVTCHVKHYTNPSQDV
TVPCPVPSTPPTPSPSTPPTPSPSCCHPRLSLHRPALEDLLLGSEANLTCT
LTGLRDASGVTFTWTPSSGKSAVQGPPDRDLCGCYSVSSVLSGCAEPW
NHGKTFTCTAAYPESKTPLTATLSKSGNTFRPEVHLLPPPSEELALNEL
VTLTCLARGFSPKDVLVRWLQGSQELPREKYLTWASRQEPSQGTTTFA
VTSILRVAAEDWKKGDTFSCMVGHEALPLAFTQKTIDRLAGKPTHVN
VSVVMAEVDGTCY

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
179
[0475] Additional constant region sequences (human IgM1 , IgM2, IgD1,
IgAl, IgG1 , IgG3, IgEl, IgE2, kappa, and lamda; mouse IgMl, kappa, and
lambda; rabbit IgMl, kappa, and lambda; and dog IgM1) may be found at
pages 296-300 of FUNDAMENTAL IMMUNOLOGY (3d ed.), William E. Paul
(ed.), Raven Press, New York, NY (1993). Many other constant region
sequences (polynucleotide and amino acid) are know in the art and may be
used in the present invention.
EXAMPLE 9
COMBINATION RADIUMMUNOTHERAPY AND CHEMOTHERAPY
[0476] The combination of chemotherapy and anti-C35 radioimmunotherapy
was shown to be more effective at reducing tumor volume than either therapy
alone. In a first experiment, the effect of combination radioimmunotherapy
with 1311 labeled 1B3 anti-C35 monoclonal antibody and chemotherapy with 5-
FluoroUracil (FU) at 150mg/kg, together with Leucovorin (LV) at 100mg/kg
was tested in Swiss nude mice grafted with Colau.C35 tumor cells. Colau.C35
are a C35 antigen positive clone of Colau cells that were tissue culture
adapted
from a human colon carcinoma and transduced with a C35 retroviral
recombinant.
[0477] Chemotherapy was initiated on day 11 following tumor graft and
300 Ci of 131I-labeled 1B3 anti-C35 monoclonal antibody was administered
on day 14. Tumor growth was followed for up to 8 weeks.
[0478] The results in Figure 5 show inhibition of tumor growth in the
group
that received combination radioimmunotherapy and chemotherapy in
comparison to the group that received chemotherapy alone or chemotherapy
and non-radiolabeled ("cold") 1B3 anti-C35 antibody. Standard parameters of
growth inhibition were calculated for the group receiving combination
radioimmunotherapy and chemotherapy in comparison to the untreated control
group.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
180
[0479] As shown
in Figure 5, Tumor Doubling Delay (TDD) equals 3.8 (at
400 mm3 tumor volume) where TDD equals (Treated - Control{in days to the
specified volume})/TVDT and TVDT = Tumor Volume Doubling Time of
Control {during exponential growth phase}. Log Cell Kill (LCK) is defined as
TDD/3.3 = 1.15, which meets the accepted standard for an effective tumor
therapy (See, e.g., Skipper HE et al., Cancer Chemotherapy Rep. 35:1-111
(1964); Coldman AJ and Goldie JH, Mathematical Biosciences 65:291-307
(1983); and Norton L and Simon R, Cancer Treat. Rep. 61:1307-1317
(1977)).
[0480] In a second experiment, the effect of combination
radioimmunotherapy
with 131I labeled 1B3 anti-C35 monoclonal antibody and chemotherapy with
cisplatin at 2 mg/kg on day 15 and 18 was tested in Swiss nude mice grafted
with Colau.C35 tumor cells. Cispaltin was administered on days 15 and 18
following tumor graft. 300 Ci of 131I-labeled 1B3 anti-C35 monoclonal
antibody was administered on day 21. Tumor growth was followed for up to
weeks.
[0481] In the same experiment, separate groups of Swiss nude mice
grafted
with Colau.C35 tumor cells were treated with either 5-FluoroUracil (5FU) at
180mg/kg, together with Leucovorin (LV) at 120mg/kg or this same
chemotherapy regimen administered on day 18 followed by 300 Ci of 131I-
labeled 1B3 anti-C35 monoclonal antibody administered on day 21.
[0482] The results in Figure 6 show some inhibition of Colau.C35 tumor
growth in the group that received chemotherapy alone (either cisplatin or
5FU/LV), greater inhibition in the group that received 131I-labeled 1B3 anti-
C35 monoclonal antibody, and even greater inhibition of tumor growth in the
group that received combination chemotherapy and 131I-labeled 1B3 anti-C35
monoclonal antibody. See Table 5, below.
=

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
181
[0483]
Table 5: Comparison of Effects of Therapeutic Modalities on Tumor Volume
in Figure 6
5FU/LV Cisplatin RIT: 5FU/LV + Cisplatin
alone alone 1311-1B3 RIT + RIT
T-C
(days) 5 5 29 37 40
TDD 0.66 0.66 3.84 4.89 5.29
LCK 0.20 0.20 1.16 1.48 1.60
RIT = radioimmunotherapy
T-C = difference in time for treated (T) and control (C) tumors to reach a
given volume (1200 mm3)
TDD = Tumor doubling delay = T-C/tumor volume doubling time of
untreated
LCK = Log Cell Kill = TDD/3.32
[0484] For convenience of use in these experimental models, a tumor
xenograft was selected that grew relatively rapidly and had to be transduced
with recombinant C35. However, the success of combination therapy was not
due to abnormally high levels of C35 expression in the transduced tumor.
Figure 7 shows that C35 expression was very similar in tumors such as 21MT1
that naturally express C35, and in C35-transduced tumors such as Colau.C35
and MDA231.C35. Cells were stained with Alexa-647 conjugated anti-C35
MAb 1F2 or isotype control. "MFI X" is the ratio of the mean fluorescence
intensity of 1F2/mean fluorescence intensity of isotype control. Hi 6N2,
derived from normal breast epithelium, and MDA_MB231, a breast tumor, and
Colau, a colon tumor, express low basal levels of C35. 21MT1, derived from
breast carcinoma, naturally expresses high levels of C35. Colau and MDA231
were transduced with empty vector (null) or human C35 recombinant vector.
All tumors were grown in vivo, tumors were excised, dissociated and stained.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
182
EXAMPLE 10
DETERMINATION OF MAXIMUM TOLERATED DOSE (MTD) OF A
CHEMOTHERAPEUTIC AGENT
[0485] Because of concerns regarding cumulative dose-limiting bone marrow
toxicity when combining chemotherapy and radioimmunotherapy, it is
necessary to determine the Maximum Tolerated Dose (MTD) of combination
therapy and, where toxicity of the two therapeutic agents is additive, adopt
strategies that will permit administration of both toxic agents. MTD is
established by regulatory criteria related to the time required for platelet
and
white cell recovery in peripheral circulation. Such standards are familiar to
those skilled in the art. In the case of murine models, an often employed
surrogate definition of MTD is the maximum dose that results in an average of
less than 20 % weight loss or less than 10% mortality. Currently established
MTD for the most common chemotherapeutic agents employed in standard
clinical protocols or in animal models are shown in Tables 6 to 9, below.
Table 6: Representative Chemotherapy Protocols in
Xenograft models (nude mice)
Cytotoxic Drug Maximum Tolerated Dose Dose + RIT Schedule
Fluorouracil/leucovor
in 180/120 mg/kg 1 150/100 mg/kg bolus, i.v.
i.p. every
day for 5
Oxaliplatin 5 mg/kg 3 TBD cycles
i.v. every 3
days for 2
Cisplatin 4 mg/kg 4'5 2 mg/kg cycles
i.p. every
day for 5
Irinotecan 15 mg/kg 3 TBD cycles
i.v. every 7
days for 2-
Taxol No toxicity 2 30 mg/kg 4'6

;3 cycles

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
183
Cytotoxic Drug Maximum Tolerated Dose Dose + RIT Schedule
i.v. every 7
days for 2
Cyclophosphamide No toxicity 2 175 mg/kg 4'7 cycles
i.v. every 4
days for 3
Adriamycin 10 mg/kg' 8 mg/kg cycles
i.p. every 3
days for 4
Gemcitabine No toxicity 5 120 mg/kg 5 cycles
Vinorelbine 20 mg/kg 4 TBD bolus, i.v.
External Beam delivered
Irradiation No toxicity 2 20 Gy locally
Notes
TBD: To be determined
1. MTD confirmed by present inventors. Maximum Tolerated Dose is defined
as > 20% average weight loss and/or > 10% lethality.
2. Non-toxic dose at indicated schedule.
3. Fichtner I et al. Anticancer drug response and expression of molecular
markers in early-passage xenotransplanted colon carcinomas. Eur J Can
2004. 40: 298-307.
4. Villena-Heinsen C et al. Human ovarian cancer xenografts in nude mice:
chemotherapy trials with paclitaxel, cisplatin, vinorelbine, and titanocene
dichloride. Anticancer Drugs 1998. 9: 557-563.
5. Higgins B. et al. Antitumor activity of erlotinib (OSI-774, Tarceva)
alone or
in combination in human non-small cell lung cancer tumor xenograft models.
Anti-Cancer Drugs 2004. 15: 503-512.
6. Kraeber-Bodere F. et al. Enhanced Antitumor Activity of Combined
pretargeted radioimmunotherapy and Paclitaxel in Medullary Thyroid Cancer
Xenograft. Mol Can I7zer 2002. 1: 267-274
7. Kraus-Berthier, L et al. Histology and sensitivity to anticancer Drugs
of two
human non-small cell lung carcinomas implanted in the pleural cavity of
nude mice. 2000. Clin Can Res 6:297-304.
Table 7: Representative Breast Cancer Chemotherapy Protocols
Regimen Cytotoxic Drug Dosage Schedule
AC doxorubicin 60 mg/m2 IV Repeat every 3
weeks for 4 cycles
cyclophosphamide 600 mg/m2 IV
CAF cyclophosphamide 100mg/m2/day PO Repeat every 28
on days 1-14 days for 6 cycles
doxorubicin 30 mg/ m2 IV on

CA 02548180 2006-06-01
WO 2005/055936 PCT/US2004/040573
184
Regimen Cytotoxic Drug Dosage Schedule
days 1 and 8
fluorouracil 500mg/ m2 IV on
days 1 and 8
CMF cyclophosphamide 100mg/m2/day PO Repeat every 28
on days 1-14 days for 6 cycles
methotrexate 40mg/ m2 IV on
days 1 and 8
fluorouracil 600mg/ m2 IV on
days 1 and 8
vinorelbine 30mg/ m2 IV on Repeat every 21
days 1 and 8 days for 6 ¨ 8
cycles
paclitaxel 175 mg/ m2 IV Repeat every 21
days for 6 cycles
Sources: DataMonitor Pipeline Insight: Breast Cancer June 2004;
http://www.bccancer.bc.ca
Table 8: Representative Colon Cancer Therapy Protocols
Regimen Drug Dosage Schedule
Leucovorin 20 mg/m2/day X 5 days Every 28 days X 6
(d1-5) IV prior to cycles
fluorouracil
Fluorouracil 425 mg/m2/day X 5 days
(d1-5) IV
Irinotecan 350 20 mg/m2 IV Repeat every 21
days for 2 -6 cycles
depending on
clinical benefit and
toxicity
FOLFOX Oxaliplatin 100 mg/m2IV Repeat every 14
days for a
Leucovorin 400 mg/m2 IV maximum of 12
cycles
Fluorouracil 400 mg/m2 IV bolus after
the Leucovorin, THEN
Fluorouracil 2400 mg/m2 IV over 46
hours
FOLFIRI Irinotecan 180 mg/m2 IV Repeat every 14
days for a
Leucovorin 400 mg/m2 IV maximum of 12

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
185
Regimen Drug Dosage Schedule
cycles
Fluorouracil 400 mg/m2 IV bolus after
the Leucovorin, THEN
Fluorouracil 2400 mg/m2 IV over 46
hours
Source: http://www.bccancenbc.ca
Table 9: Representative Lung Cancer Chemotherapy Protocols
Drug Dose Schedule
Docetaxel 75 mg/m2 IV Repeat every 21 days X 6
cycles
Docetaxel 75 mg/m2 IV Repeat every 21 days X 4
cycles
Cisplatin 75 mg/m2IV
Cisplatin 75 mg/m2 IV on Day 1 Repeat every 21 days X 6
cycles
Gemcitabine 1250 mg/m2 IV on Day 1
and Day 8
Source: http://www.bccancer.bc.ca
[0486] Effective strategies to reduce the combined MTD of treatment with
chemotherapy and radioimmunotherapy include: reducing the dose of
chemotherapeutic agent administered to a level which does not result in
additive toxicity when administered in conjunction with radioimmunotherapy
at its MTD. (see Example 10A, below); selecting a chemotherapeutic agent
that does not contribute additive toxicity when employed in combination with
radioimmunotherapy. (see Example 10B, below); and reducing the bone
marrow toxicity of the radioimmunotherapeutic agent (see Example 10C,
below).
A. Reducing the dose of chemotherapeutic agent to reduce toxicity.
[0487] 2 mg/kg of cisplatin (approximately 50% of MTD) was administered
to
Swiss nude mice on days 15 and 18 followed 72 hours later by 1311-labeled
1B3 anti-C35 monoclonal antibody administered at its MTD (300 [iCi). As

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
186
shown in Figure 8, the combined toxicity of cisplatin and
radioimmunotherapeutic as determined by weight loss was not significantly
different from toxicity of the radioimmunotherapeutic alone administered at
its
MTD.
B. Chemotherapeutic agent does not contribute to additive toxicity.
[0488] 5-FluoroUracil at the MTD of 180 mg/kg together with Leucovorin at
120 mg/kg were administered on day 18 followed by 131I-labeled 1B3 anti-C35
monoclonal antibody administered at its MTD (300 1.1Ci) on day 21. As
shown, in Figure 8, the MTD of the combination of the two toxic agents was
not exceeded, even though each agent was administered at its individual MTD.
C. Reduced bone marrow toxicity of the radioimmunotherapeutic agent.
[0489] An alternative strategy is to reduce the bone marrow toxicity of
the
radioimmunotherapeutic by biochemical modifications that result in
accelerated clearance of radiolabeled antibody from peripheral circulation.
Appropriate modifications include use of different antibody isotypes such as
IgG3, IgA, IgD or IgE as indicated in Table 10 or deletion of the CH2 domain
of IgG, which is responsible for its extended serum half life (See Mueller BM,

RA. Reisfeld, and SD Gillies, Proc. Natl. Acad. Sci. USA 87:5702-5705
(1990); Slavin-Chiorini DC, et al., Int. J. Cancer 53:97-103 (1993)).
Table 10: Serum Half-Life of Human Immunoglobulin Isotypes
Immunoglobulin Isotype Serum Half-Life
IgG1 21 days
IgG2 20 days
IgG3 7 days
IgG4 21 days
IgM 10 days

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
187
Immunoglobulin Isotype Serum Half-Life
IgA 6 days
IgD 3 days
IgE 2 days
[0490] Other strategies to engineer antibodies and/or antibody fragments,
or
otherwise modify antibody structure so as to reduce serum half-life are also
applicable. Examples of such engineered antibodies and/or antibody
fragments include, but are not limited to, domain-deleted antibodies, Fab,
F(ab')2, scFv, minibodies, diabodies, triabodies, tetrabodies, etc.
EXAMPLE 11
C35 PEPTIDE EPITOPES OF 1B3 AND 1F2 ANTIBODIES
[0491] To localize the epitope specificity of 1B3 and 1F2 antibodies,
recombinant human C35 (rhC35) synthesized with a 6x His tag in E. coli was
digested with Lys-C endoproteinase. This enzyme cuts after lysine (K)
residues in the protein sequence. Figure 9 shows the expected peptide
fragments following complete digestion of rhC35 with Lys-C. The full
sequence of rhC35, including the amino terminal 6x His tag addition is shown.
Amino acid positions are numbered relative to the amino terminal methionine
(M) of the native human sequence. Note that digestion at the first and third
lysine followed by negatively charged residues is inefficient and some longer
combination fragments may be generated. Lys-C endoproteinase was added to
purified rhC35 at a 50:1 weight ratio and incubated for 18 hours at 37 C in
25mM Tris, pH 8Ø The digest was ethanol precipitated and resolubilized in
reducing Tricine sample buffer. After heating 5 minutes at 100 C, samples
were separated by electrophoresis on a 16% Tricine gel (Invitrogen). Peptides
were transferred to PVDF membranes and the blots depicted in Figure 10 were
either stained with Coomassie blue (lanes 1-3 of both left and right panels)
to

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
188
detect all peptides or processed with one of the following: 1 1.1,g/m1 of the
murine 1F2 anti-C35 antibody followed by alkaline phosphatase conjugated
goat anti-mouse antibody (lane 4 of left panel); 1 pg/m1 of the 1B3 anti-C35
immunoglobulin variable regions linked to human constant regions (MAbll)
followed by alkaline phosphatase conjugated goat anti-human antibody (lane 5
of left panel); or anti-6x His tag mouse antibody (Amersham) followed by
alkaline phosphatase conjugated goat anti-mouse antibody (lane 4 and 5 of
right panel). BCIP/NBT substrate was added and developed to detect the
secondary reagents. Molecular weight markers are indicated in the flanking
lanes of both panels.
[0492] In Figure 10, the indicated band A migrates at the position of
undigested rhC35. Note that band B stains with 1F2 but not MAbll (1B3)
anti-C35 antibody while band C stains with neither 1F2 nor MAbll (1B3)
antibody. Since bands B and C both do stain with anti-6x His tag antibody to
the 6x His tag at the amino terminus of rhC35, it may be concluded that both
fragments lack C-terminal peptide fragments. In the case of the approximately
15kDa band 11_3, the missing epitope required for staining with MAbll (1B3) is

the 11 amino acid C-terminal C35 peptide ITNSRPPCVIL representing
residues 105-115 of the native C35 sequence. This epitope is not required for
staining with 1F2. In contrast, the 1F2 antibody does not react with the
approximately SkDa band C which is, in addition, lacking residues 53-104 of
the native C35 sequence. The
results demonstrate that 1B3 antibody is
specific for an epitope within C35 residues 105-115 (ITNSRPPCVIL),
whereas 1F2 antibody is specific for an epitope within the C35 residues 53-
104.

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
189
EXAMPLE 12
HUMAN ANTIBODIES RELATED TO 1B3 ANTI-C35
MONOCLONAL ANTIBODY
[0493] Two antibodies which are human-derived except for having the same
immunoglobulin heavy chain CDR3 region as the mouse 1B3 anti-C35
monoclonal antibody were generated by the method disclosed in US 2002
0123057 Al, published 5 September 2002.
[0494] MAb 165 comprises the 141D10 VH H732 heavy chain variable region
(SEQ ID NOS: 56 and 57), and the UH8 VK L120 kappa light chain variable
region (SEQ ID NOS: 58 and 59). As shown in Figure 11, MAb 165 is C35-
specific. 141D10 recombinant vaccinia virus was co-infected into HeLa cells
with UH8 recombinant vaccinia virus. The resulting secreted antibody was
tested for binding to C35 or control protein A27L (vaccinia virus protein) by
ELISA.
[0495] MAb 171 comprises the MSH3 VH H835 heavy chain variable region
(SEQ ID NOS:60 and 61) and the UH8 VK L120 kappa light chain variable
region (SEQ ID NOS: 58 and 59).
EXAMPLE 13
IDENTIFICATION OF C35 PEPTIDE EPITOPES RECOGNIZED BY
ANTI-C35 ANTIBODIES
[0496] Rabbit polyclonal antibodies were raised to recombinant C35
employing standard immunization methods well known in the art.
Overlapping peptides 15 amino acids in length were synthesized
corresponding to the 115 amino acid long C35 protein sequence beginning at
each amino acid residue from 1 to 101. Peptides are named based on the C35
position of the amino terminus residue in each 15 amino acid peptide. In each
peptide that overlapped the cysteine residues at positions 30, 33 and 112 of
the

CA 02548180 2011-12-28
190
natural C35 sequence, alanine was substituted for cysteine to avoid formation
of disulfide crosslinked peptides.
[0497] Wells of 96 well MaxisorTMp microtiter plates were coated with
either 2
pg C35 protein, or 14 lig or 40 pg of the indicated 15 amino acid peptide
derived from the C35 protein sequence and binding of antibodies in the rabbit
C35 immune serum was determined as described in detail below. Data in
Table 11, below, is shown for the positive and negative controls and for those

C35 derived peptides for which positive binding was detected. Variations in
the level of peptide binding can be due to either differences in the
concentration of specific antibody species, differences in antibody affinity
or a
combination thereof.
A. Peptide sample preparation
[0498] C35 peptide 15-mers in 100% DMSO, 10mg/ml, were aliquotted into
1.5 ml tubes, 40 g/tube, under sterile conditions, then speed vacuumed to
remove DMSO. The peptides were resuspended in PBS, p117.2, lml/tube,
mixed well and spun down. Each peptide concentration ("peptide solution")
was 40 g/ml. Peptide solution should be stored at ¨20 C until use. Once
thawed, it should be kept at 4 C for no more than 2 weeks.
B. Coating samples on the Maxisorp plates
[0499] For 14 1.1g/m1 peptide coated plates, 65 I PBS, pH7.2 per well was
added, followed by 35 1 peptide solution per well (for a total volume of 100
l/well), and mixed well. For 40 g/m1 peptide coated plates, 100111 of peptide

solution was placed directly on the plate, 100 1/well. Control C35 protein was

diluted into PBS, pH7.2 to 2 g/m1 and added to the plates, 100 l/well.
Coated plates were incubated at room temperature for 2 hours, then 4 C
overnight.

CA 02548180 2006-06-01
WO 2005/055936 PCT/US2004/040573
191
C. ELISA conditions
[0500] Each plate was washed 3 times on a plate washer. Plates were
blocked
with "blocking buffer" (PBS, pH7.2 and 10% FBS) at room temperature for 2
hours, followed by washing each plate 3 times on plate washer. The primary
antibody, rabbit anti-human C35 polyclonal antibody (made by Bethyl, 62902
batch) was diluted into "assay diluent" (PBS, pH7.2 plus 0.05% Tween 20 and
10% FBS) and added to the plates, 100 1/well. Plates were incubated at room
temperature for 2 hours. For 14 ,g/m1 peptide coated plates, 100 ng/ml of
primary antibody was added. For 40 ps/m1 peptide coated plates, 1 jig/m1 of
primary antibody was added. Plates were washed 5 times on a plate washer.
The secondary antibody, HRP (horseradish peroxidase conjugated goat anti-
rabbit Fc polyclonal antibody from Zymed, was diluted at a 1:20,000 dilution
into assay diluent and added to the plates, 100 1/well. The plates were
incubated at room temperature for 2 hours. Plates were washed 7 times on a
plate washer. Substrate was added as per the kit manufacturers instructions
and incubated at room temperature in the dark for 15 minutes. The reaction
was stopped by adding 2N H2SO4, 100 1.11/well. Absorbance at 450-570 nm
was read immediately.
Table 11: Antibody Binding to C35 Epitopes
Absorbance @ 450-570 nm
Peptide Sequence 14 tg/m1 40 tg/m1
None 0.009 0.013
without
primary rabbit C35 protein or peptides 0.009 0.009
antibody
C35 protein C35 protein 3.765 3.5225
P1 MSGEPGQTSVAPPPE 0.257
P2 SGEPGQTSVAPPPEE 0.108 1.227
P3 GEPGQTSVAPPPEEV 1.458 3.349

CA 02548180 2006-06-01
WO 2005/055936 PCT/US2004/040573
192
Peptide Sequence 14 gig/m1 40 pg/m1
P4 EPGQTSVAPPPEEVE 1.254 3.282
P5 PGQTSVAPPPEEVEP 2.719 3.383
P6 GQTSVAPPPEEVEPG 2.483 3 .381
P7 QTSVAPPPEEVEP GS 2.635 3.388
P8 T SVAPPPEEVEP GS G 0.059 0.394
P9 SVAPPPEEVEPGSGV 2.31 3.367
P10 VAPPPEEVEP GS GVR 1.736 3.407
P11 APPPEEVEPGSGVRI 1.526 3.317
P12 PPPEEVEPGSGVRTV 0.836
P13 PPEEVEP GS GVRIVV 0.385 2.522
P14 PEEVEPGSGVRIVVE 0.127 0.972
P15 EEVEPGSGVRIVVEY 0.039 0.334
P16 EVEPGSGVRIVVEYA 0.027 0.172
P62 TGAFEIEINGQLVFS 0.023 0.107
P63 GAFEIEINGQLVFSK 0.079 0.557
P64 AFEIEINGQLVFSKL 0.055 0.423
P65 FEIEINGQLVFSKLE 0.043 0.269
P66 EIEINGQLVFSKLEN 0.028 0.182
P80 NGGFPYEKDLIEMR. 0.018 0.1
P81 GGFPYEKDLIEAIRR 0.032 0.204
P82 GFPYEKDLIEAIRRA 0.02 0.18
P83 FPYEKDLIEAIRRAS 0.025 0.19
P84 PYEKDLIEAIRRASN 0.037 0.391
P85 YEKDLIEMRRASNG 0.024 0.179
P86 EKDLTEAIRRASNGE 0.023 0.166
P88 DLIEAIRRASNGETL 0.025 0.135
P89 LIEAIRRASNGETLE 0.015 0.053
P90 IEMRRASNGETLEK 0.04 0.403

CA 02548180 2011-12-28
193
Peptide - Sequence 14 pg/m1 40
P91 EAERRASNGETLEKI 0.023 0.144
P92 AIRRASNGETLEKIT 0.04 0.267
P93 IRRASNGETLEKITN 0.051 0.389
P94 RRASNGETLEKITNS 0.061 0.526
P95 RASNGETLEKTINSR 0.062 0.462
P97 SNGETLEKITNSRPP 0.046 0.373
P98 NGETLEKITNSRPPA 0.035 0.213
P99 GETLEKITNSRPPAV 0.026 0.18
P100 ETLEKITNSRPPAVI 0.017 0.124
P101 TLEKITNSRPPAVIL 0.472 2.519
[0501]

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
194
_
_______________________________________________________________________________
_
1500tint'iijgeithfiue International application To Be
Assigned
reference number 1843.019PC03
INDICATIONS RELATING TO A DEPOSITED MICROORGANISM
(PCT Rule 13bis)
A. The indications made below relate to the microorganism referred to in the
description on page 33 , line
17,18
B. IDENTIFICATION OF DEPOSIT
Further deposits are identified on an additional sheet El
Name of depositary institution
American Type Culture Collection
Address of depositary institution (including postal code and country)
10801 University Boulevard
Manassas, Virginia 20110
United States of America
Date of deposit Accession Number
November 11, 2003 PTA-5637
C. ADDITIONAL INDICATIONS (leave blank if not applicable) This information
is continued on an additional sheet 0
Plasmid DNA: 1B3G
= D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE (if the indications
are not for all designated States)
=
E. SEPARATE FURNISHING OF INDICATIONS (leave blank If not applicable)
The indications listed below will be submitted to the international Bureau
later (specifr the general nature of the indications, e.g.,
"Accession Number of Deposit")
=
=:,
For receiving Office use only __________________ For International Bureau
use only
is sheet was 'received with the international application 0 This sheet was
received by the International Bureau on:
FirrAR DEC no4
Authorized offirolanda V. Harrod Authorized officer
PCT/Internat'l Appl Processing Div
043) aQ5-0079, ______________ J6--j
Form PCT/110/134 (July 1992)
#205566v1<SKGF_D01> -Deposit 1..wpd

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
195
Mill-6min or ant' S tile International application To Be
Assigned
reference number 1843.019PC03
INDICATIONS RELATING TO A DEPOSITED MICROORGANISM
(PCT Rule 13bis)
A. The indications made below relate to the microorganism referred to in the
description on page 33 , line
18, 19, 20
B. IDENTIFICATION OF DEPOSIT Further deposits are identified on an
additional sheet El
Name of depositary institution
American Type Culture Collection
Address of depositary institution (including postal code and country)
10801 University Boulevard
Manassas, Virginia 20110
United States of America
Date of deposit Accession Number
November 11,2003 PTA-5638
C. ADDITIONAL INDICATIONS (leave blank if not applicable) This information
is continued on an additional sheet El
Plasmid DNA: 1B3K
D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE (if the indications are
not for all designated States)
E. SEPARATE FURNISHING OF INDICATIONS (leave blank ff not applicable)
The indications listed below will be submitted to the international Bureau
later (specifr the general nature of the indications, e.g.,
"Accession Number of Deposit")
=
For receiving Office use only _______ k ________ For International Bureau
use only
Ull4i-s sheet was received with the internationala lic 0 This sheet was
received by the International Bureau on:
rip oir.!, 0 Dt't ttit
Authorized oitimtlnda V Harrod Authorized officer
PCT/internaVi Appl Processing Div
(703) 3Q5-3670
Form PDT/RD/134 (July 1992)
#205571v1<SKGF DC1> -Deposit 2.vvpd

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
196
Apfolidarit'S'or agtrimar International application To Be
Assigned
reference number 1843.019PC03
INDICATIONS RELATING TO A DEPOSITED MICROORGANISM
(PCT Rule 13bis)
A. The indications made below relate to the microorganism referred to in the
description on page _ 33 , line
14,15
B. IDENTIFICATION OF DEPOSIT
Further deposits are identified on an additional sheet 0
Name of depositary institution
American Type Culture Collection
Address of depositary institution (including postal code and country)
10801 University Boulevard
Manassas, Virginia 20110
United States of America
Date of deposit Accession Number
November 11, 2003 PTA-5639
C. ADDITIONAL INDICATIONS (leave blank if not applicable)
This information is continued on an additional sheet 0
Plasmid DNA: 1F2G
D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE Wale indications are not
for all designated States)
E. SEPARATE FURNISHING OF INDICATIONS (leave blank if noi applicable)
The indications listed below will be submitted to the international Bureau
later (spec05, the general nature of the indications, e.g.,
"Accession Number of Deposit")
=
For receiving Office use only __________________ For International Bureau
use only
13-1<sheet was regved with the inte atnal.ap 0 This sheet was received by
the International Bureau on:
Authorized officer Authorized officer
Yolanda V. Harrod
PCT/Internarl Appl Processing
CIP1) '4115-'70 ____________________
Form PCT/R0/134 (July 1992)
#205576v1<SICOF Del> -Deposit 3.1vpd

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
197
Arlicbnts oragents tile International application. - To :Be
Assigned
reference number 1-843.019PC03
INDICATIONS RELATING TO A DEPOSITED MICROORGANISM
(PCT Rule 13bis)
A. The indications made below relate to the microorganism referred to in the
description on page 3 , line
15 16 17
B. IDENTIFICATION OF DEPOSIT Further deposits are identified on an
additional sheet El
Name of depositary institution
American Type Culture Collection
Address of depositary institution (including postal code and countly)
10801 University Boulevard
Manassas, Virginia 20110
United States of America
Date of deposit Accession Number
November 11, 2003 PTA-5640
C. ADDITIONAL INDICATIONS (leave blank If not applicable) This information
is continued on an additional sheet 0
Plasmid DNA: 1F1K
D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE of the indications are not
for all designated States)
E. SEPARATE FURNISHING OF INDICATIONS (leave blank if nol applicable)
The indications listed below will be submitted to the international Bureau
later (specifi) the general nature of the indications, e.g.,
"Accession Number of Deposit")
_________________________________________________ For receiving Office use
only For International Bureau use only
Ijiµts sheet was rdFived with the internatio 1 p firtm 0 This sheet was
received by the International Bureau on:
aJt4
e
Ns j zo =
Authorized offiGlanda V. Harrod Authorized officer
PCT/internarl App l Processing Div
(703) 305-3670 = 314_,
Form PCT/RO/134 (July 1992) #205578v1<SK0F
DC1> -Deposit 4.wpd

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
198
;-1
Applicant's or agent's file International application To Be
Assigned
reference number 1843.019P'CO3
INDICATIONS RELATING TO A DEPOSITED MICROORGANISM
(PCT Rule 13bis)
A. The indications made below relate to the microorganism referred to in the
description on page 40 , line
28,29
B. IDENTIFICATION OF DEPOSIT
Further deposits are identified on an additional sheet '2
Name of depositary institution
American Type Culture Collection =
Address of depositary institution (including postal code and country)
10801 University Boulevard
Manassas, Virginia 20110
United States of America
Date of deposit Accession Number
November 11, 2003 PTA-5641
C. ADDITIONAL INDICATIONS (leave blank if not applicable)
This information is continued on an additional sheet 0
Plasmid DNA: H0009
D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE (if the indications are
not for all designated States)
E. SEPARATE FURNISHING OF INDICATIONS (leave blank if not applicable)
The indications listed below will be submitted to the international Bureau
later (spectfr the general nature of the indications, e.g.,
'Accession Number of Deposit')
_________________________________________________ For receiving Office use
only For International Bureau use only
This sheet was received with the internatiftal apAcition 0 This sheet was
received by the International Bureau on:
VIVO 1147 '0 6 .DEe Z
Authorized offiocetand v. Harrod
r Authorized officer
ya
PCT/Internari Appi Proowdng D'y
(703) 305-3670
Form PCT/RO/134 (July 1992) #205579v1<SKGF DC1> -
Deposit 5-DNA 1.wpd

CA 02548180 2006-06-01
WO 2005/055936
PCT/US2004/040573
199
Applicants or agents file International applicatior To Be
Assigned
reference number 1843.019PC03
INDICATIONS RELATING TO A DEPOSITED MICROORGANISM
(PCT Rule 13bis)
A. The indications made below relate to the microorganism referred to in the
description on page _ 40 , line
29,30
B. IDENTIFICATION OF DEPOSIT Further deposits are identified on an
additional sheet 0
Name of depositary institution
American Type Culture Collection
Address of depositary institution (including postal code and country)
10801 University Boulevard
Manassas, Virginia 20110
United States of America
Date of deposit Accession Number
November 11, 2003 PTA-5642
C. ADDITIONAL INDICATIONS (leave blank if not applicable) This information
is continued on an additional sheet 0
Plasmid DNA: L0010
D. DESIGNATED STATES FOR WHICH INDICATIONS ARE MADE (( the indications are not
for all designated States)
E. SEPARATE FURNISHING OF INDICATIONS (leave blank if not applicable)
The indications listed below will be submitted to the international Bureau
later (specifir the general nature of the indications, e.g.,
"Accession Number of Deposit")
=
For receiving Office use only ___________________ For International Bureau
use only
Wrhis sheet was received with the international application 0 This sheet
was received by the International Bureau on:
R:* DEC ?MN
Authorized offic*bianda V. Harrod
Authorized officer
(P7C0 3T) p
/ I 3n toet in3aet 7' I . pl Processing Dicp,
Form PCT/RO/134 (July 1992)
4205582v1<SKGF DC1> -Deposit 6-DNA 2.wpd

DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PLUS D'UN TOME.
CECI EST LE TOME 1 DE 2
NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des
Brevets.
JUMBO APPLICATIONS / PATENTS
THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.
THIS IS VOLUME 1 OF 2
NOTE: For additional volumes please contact the Canadian Patent Office.

Representative Drawing

Sorry, the representative drawing for patent document number 2548180 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2014-02-04
(86) PCT Filing Date 2004-12-06
(87) PCT Publication Date 2005-06-23
(85) National Entry 2006-06-01
Examination Requested 2009-11-30
(45) Issued 2014-02-04
Deemed Expired 2019-12-06

Abandonment History

There is no abandonment history.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2006-06-01
Maintenance Fee - Application - New Act 2 2006-12-06 $100.00 2006-06-01
Registration of a document - section 124 $100.00 2006-09-22
Registration of a document - section 124 $100.00 2006-09-22
Registration of a document - section 124 $100.00 2006-09-22
Maintenance Fee - Application - New Act 3 2007-12-06 $100.00 2007-09-25
Maintenance Fee - Application - New Act 4 2008-12-08 $100.00 2008-09-25
Request for Examination $800.00 2009-11-30
Maintenance Fee - Application - New Act 5 2009-12-07 $200.00 2009-11-30
Maintenance Fee - Application - New Act 6 2010-12-06 $200.00 2010-12-02
Maintenance Fee - Application - New Act 7 2011-12-06 $200.00 2011-12-06
Maintenance Fee - Application - New Act 8 2012-12-06 $200.00 2012-11-20
Registration of a document - section 124 $100.00 2013-08-29
Final Fee $1,152.00 2013-10-24
Maintenance Fee - Application - New Act 9 2013-12-06 $200.00 2013-11-28
Maintenance Fee - Patent - New Act 10 2014-12-08 $250.00 2014-12-01
Maintenance Fee - Patent - New Act 11 2015-12-07 $250.00 2015-11-30
Maintenance Fee - Patent - New Act 12 2016-12-06 $250.00 2016-12-05
Maintenance Fee - Patent - New Act 13 2017-12-06 $250.00 2017-12-04
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
VACCINEX, INC.
Past Owners on Record
EVANS, ELIZABETH E.
PARIS, MARK J.
SAHASRABUDHE, DEEPAK M.
SMITH, ERNEST S.
ZAUDERER, MAURICE
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2006-09-22 201 10,639
Description 2006-09-22 27 751
Abstract 2006-06-01 1 61
Claims 2006-06-01 18 464
Drawings 2006-06-01 11 751
Description 2006-06-01 201 10,639
Description 2006-06-01 27 756
Cover Page 2006-08-16 1 32
Description 2011-12-28 201 10,271
Description 2011-12-28 27 751
Claims 2011-12-28 6 222
Claims 2012-12-18 7 229
Cover Page 2014-01-08 1 32
PCT 2006-06-01 2 70
Assignment 2006-06-01 3 108
Correspondence 2006-08-14 1 27
Assignment 2006-09-22 14 523
Prosecution-Amendment 2006-09-22 3 83
Prosecution-Amendment 2009-11-30 2 62
Prosecution-Amendment 2011-06-29 4 182
Prosecution-Amendment 2011-12-28 54 2,468
Prosecution-Amendment 2012-06-27 2 64
Prosecution-Amendment 2012-12-18 13 426
Assignment 2013-08-29 12 320
Correspondence 2013-10-24 2 65

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :